1
|
Xu N, Chen SY, Tang AH. Tuning synapse strength by nanocolumn plasticity. Trends Neurosci 2025; 48:200-212. [PMID: 39848836 DOI: 10.1016/j.tins.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/25/2025]
Abstract
The precise organization of the complex set of synaptic proteins at the nanometer scale is crucial for synaptic transmission. At the heart of this nanoscale architecture lies the nanocolumn. This aligns presynaptic neurotransmitter release with a high local density of postsynaptic receptor channels, thereby optimizing synaptic strength. Although synapses exhibit diverse protein compositions and nanoscale organizations, the role of structural diversity in the notable differences observed in synaptic physiology remains poorly understood. In this review we examine the current literature on the molecular mechanisms underlying the formation and maintenance of nanocolumns, as well as their role in modulating various aspects of synaptic transmission. We also discuss how the reorganization of nanocolumns contributes to functional dynamics in both synaptic plasticity and pathology.
Collapse
Affiliation(s)
- Na Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; School of Medicine, Anhui University of Science and Technology, Huainan 232001, China.
| | - Si-Yu Chen
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Ai-Hui Tang
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China.
| |
Collapse
|
2
|
Pollitt SL, Levy AD, Anderson MC, Blanpied TA. Large Donor CRISPR for Whole-Coding Sequence Replacement of Cell Adhesion Molecule LRRTM2. J Neurosci 2025; 45:e1461242024. [PMID: 39824639 PMCID: PMC11823385 DOI: 10.1523/jneurosci.1461-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/01/2024] [Accepted: 12/02/2024] [Indexed: 01/20/2025] Open
Abstract
The cell adhesion molecule leucine-rich repeat transmembrane neuronal protein 2 (LRRTM2) is crucial for synapse development and function. However, our understanding of its endogenous trafficking has been limited due to difficulties in manipulating its coding sequence (CDS) using standard genome editing techniques. Instead, we replaced the entire LRRTM2 CDS by adapting a two-guide CRISPR knock-in method, enabling complete control of LRRTM2. In primary rat hippocampal cultures dissociated from embryos of both sexes, N-terminally tagged, endogenous LRRTM2 was found in 80% of synapses, and synaptic LRRTM2 content correlated with PSD-95 and AMPAR levels. LRRTM2 was also enriched with AMPARs outside synapses, demonstrating the sensitivity of this method to detect relevant new biology. Finally, we leveraged total genomic control to increase the synaptic levels of LRRTM2 via simultaneous mutation of its C-terminal domain, which did not correspondingly increase AMPAR enrichment. The coding region of thousands of genes span lengths suitable for whole-CDS replacement, suggesting this simple approach will enable straightforward structure-function analysis in neurons.
Collapse
Affiliation(s)
- Stephanie L Pollitt
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Michael C Anderson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| |
Collapse
|
3
|
Zhao G, Sathkumara HD, Miranda-Hernandez S, Seifert J, Valencia-Hernandez AM, Puri M, Huang W, Toth I, Daly N, Skwarczynski M, Kupz A. A Modular Self-Assembling and Self-Adjuvanting Multiepitope Peptide Nanoparticle Vaccine Platform to Improve the Efficacy and Immunogenicity of BCG. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406874. [PMID: 39757706 DOI: 10.1002/smll.202406874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/29/2024] [Indexed: 01/07/2025]
Abstract
After more than a century since its initial development, Bacille Calmette-Guérin (BCG) remains the only licensed vaccine against tuberculosis (TB). Subunit boosters are considered a viable strategy to enhance BCG efficacy, which often wanes in adolescence. While many studies on booster subunit vaccines have concentrated on recombinant proteins, here we developed a novel modular peptide-based subunit vaccine platform that is flexible, cold-chain independent and customizable to diverse circumstances and populations. Each individual peptide building block consists of a linear arrangement comprising a 15-leucine self-assembly inducer moiety, a Mycobacterium tuberculosis (Mtb) target epitope and an human leukocyte antigen E (HLA-E) binding moiety, with each moiety separated by a triple lysine spacer. The building blocks, in any combination, are able to form a multiepitope nanoparticle. Six Mtb epitopes were selected to produce the self-assembling and self-adjuvating peptide-based TB nano-vaccine candidate PNx6. In vivo vaccination-challenge experiments demonstrated that subcutaneous boost of parenteral BCG immunization with PNx6 significantly enhanced its immunogenicity and improved its protective efficacy in a murine model of TB by more than 5-fold. This study presents evidence that purely amphiphilic peptides self-assemble into self-adjuvating nanoparticles with appropriate size and morphology for TB vaccination with great potential for a multitude of other diseases.
Collapse
Affiliation(s)
- Guangzu Zhao
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Harindra D Sathkumara
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Socorro Miranda-Hernandez
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Julia Seifert
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Ana Maria Valencia-Hernandez
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Munish Puri
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Wenbin Huang
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Istvan Toth
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Norelle Daly
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| | - Mariusz Skwarczynski
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Andreas Kupz
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns & Townsville, QLD, 4878 & 4811, Australia
| |
Collapse
|
4
|
Liouta K, Lubas M, Venugopal V, Chabbert J, Jeannière C, Diaz C, Munier M, Tessier B, Claverol S, Favereaux A, Sainlos M, de Wit J, Letellier M, Thoumine O, Chamma I. LRRTM2 controls presynapse nano-organization and AMPA receptor sub-positioning through Neurexin-binding interface. Nat Commun 2024; 15:8807. [PMID: 39394199 PMCID: PMC11470055 DOI: 10.1038/s41467-024-53090-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
Synapses are organized into nanocolumns that control synaptic transmission efficacy through precise alignment of postsynaptic neurotransmitter receptors and presynaptic release sites. Recent evidence show that Leucine-Rich Repeat Transmembrane protein LRRTM2, highly enriched and confined at synapses, interacts with Neurexins through its C-terminal cap, but the role of this binding interface has not been explored in synapse formation and function. Here, we develop a conditional knock-out mouse model (cKO) to address the molecular mechanisms of LRRTM2 regulation, and its role in synapse organization and function. We show that LRRTM2 cKO specifically impairs excitatory synapse formation and function in mice. Surface expression, synaptic clustering, and membrane dynamics of LRRTM2 are tightly controlled by selective motifs in the C-terminal domain. Conversely, the N-terminal domain controls presynapse nano-organization and postsynapse AMPAR sub-positioning and stabilization through the recently identified Neurexin-binding interface. Thus, we identify LRRTM2 as a central organizer of pre- and post- excitatory synapse nanostructure through interaction with presynaptic Neurexins.
Collapse
Affiliation(s)
- Konstantina Liouta
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Malgorzata Lubas
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Vasika Venugopal
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Julia Chabbert
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Caroline Jeannière
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Candice Diaz
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Matthieu Munier
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Béatrice Tessier
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | | | - Alexandre Favereaux
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Matthieu Sainlos
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven Brain Institute, Leuven, Belgium
| | - Mathieu Letellier
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Olivier Thoumine
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Ingrid Chamma
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
5
|
Stokes EG, Kim H, Ko J, Aoto J. A Systematic Structure-Function Characterization of a Human Mutation in Neurexin-3α Reveals an Extracellular Modulatory Sequence That Stabilizes Neuroligin-1 Binding to Enhance the Postsynaptic Properties of Excitatory Synapses. J Neurosci 2024; 44:e1847232024. [PMID: 39231636 PMCID: PMC11466067 DOI: 10.1523/jneurosci.1847-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024] Open
Abstract
α-Neurexins are essential and highly expressed presynaptic cell-adhesion molecules that are frequently linked to neuropsychiatric and neurodevelopmental disorders. Despite their importance, how the elaborate extracellular sequences of α-neurexins contribute to synapse function is poorly understood. We recently characterized the presynaptic gain-of-function phenotype caused by a missense mutation in an evolutionarily conserved extracellular sequence of neurexin-3α (A687T) that we identified in a patient diagnosed with profound intellectual disability and epilepsy. The striking A687T gain-of-function mutation on neurexin-3α prompted us to systematically test using mutants whether the presynaptic gain-of-function phenotype is a consequence of the addition of side-chain bulk (i.e., A687V) or polar/hydrophilic properties (i.e., A687S). We used multidisciplinary approaches in mixed-sex primary hippocampal cultures to assess the impact of the neurexin-3αA687 residue on synapse morphology, function and ligand binding. Unexpectedly, neither A687V nor A687S recapitulated the neurexin-3α A687T phenotype. Instead, distinct from A687T, molecular replacement with A687S significantly enhanced postsynaptic properties exclusively at excitatory synapses and selectively increased binding to neuroligin-1 and neuroligin-3 without changing binding to neuroligin-2 or LRRTM2. Importantly, we provide the first experimental evidence supporting the notion that the position A687 of neurexin-3α and the N-terminal sequences of neuroligins may contribute to the stability of α-neurexin-neuroligin-1 trans-synaptic interactions and that these interactions may specifically regulate the postsynaptic strength of excitatory synapses.
Collapse
Affiliation(s)
- Eric G Stokes
- Department of Pharmacology, University of Colorado Anschutz School of Medicine, Colorado 80045
| | - Hyeonho Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, Korea
| | - Jason Aoto
- Department of Pharmacology, University of Colorado Anschutz School of Medicine, Colorado 80045
| |
Collapse
|
6
|
Bai SY, Zeng DY, Ouyang M, Zeng Y, Tan W, Xu L. Synaptic cell adhesion molecules contribute to the pathogenesis and progression of fragile X syndrome. Front Cell Neurosci 2024; 18:1393536. [PMID: 39022311 PMCID: PMC11252757 DOI: 10.3389/fncel.2024.1393536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and a monogenic cause of autism spectrum disorders. Deficiencies in the fragile X messenger ribonucleoprotein, encoded by the FMR1 gene, lead to various anatomical and pathophysiological abnormalities and behavioral deficits, such as spine dysmorphogenesis and learning and memory impairments. Synaptic cell adhesion molecules (CAMs) play crucial roles in synapse formation and neural signal transmission by promoting the formation of new synaptic contacts, accurately organizing presynaptic and postsynaptic protein complexes, and ensuring the accuracy of signal transmission. Recent studies have implicated synaptic CAMs such as the immunoglobulin superfamily, N-cadherin, leucine-rich repeat proteins, and neuroligin-1 in the pathogenesis of FXS and found that they contribute to defects in dendritic spines and synaptic plasticity in FXS animal models. This review systematically summarizes the biological associations between nine representative synaptic CAMs and FMRP, as well as the functional consequences of the interaction, to provide new insights into the mechanisms of abnormal synaptic development in FXS.
Collapse
Affiliation(s)
- Shu-Yuan Bai
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - De-Yang Zeng
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - Ming Ouyang
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - Yan Zeng
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - Wei Tan
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - Lang Xu
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
George K, Hoang HT, Tibbs T, Nagaraja RY, Li G, Troyano-Rodriguez E, Ahmad M. Robust GRK2/3/6-dependent desensitization of oxytocin receptor in neurons. iScience 2024; 27:110047. [PMID: 38883814 PMCID: PMC11179071 DOI: 10.1016/j.isci.2024.110047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/22/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Oxytocin plays critical roles in the brain as a neuromodulator, regulating social and other affective behavior. However, the regulatory mechanisms controlling oxytocin receptor (OXTR) signaling in neurons remain unexplored. In this study, we have identified robust and rapid-onset desensitization of OXTR response in multiple regions of the mouse brain. Both cell autonomous spiking response and presynaptic activation undergo similar agonist-induced desensitization. G-protein-coupled receptor kinases (GRK) GRK2, GRK3, and GRK6 are recruited to the activated OXTR in neurons, followed by recruitment of β-arrestin-1 and -2. Neuronal OXTR desensitization was impaired by suppression of GRK2/3/6 kinase activity but remained unaltered with double knockout of β-arrestin-1 and -2. Additionally, we observed robust agonist-induced internalization of neuronal OXTR and its Rab5-dependent recruitment to early endosomes, which was impaired by GRK2/3/6 inhibition. This work defines distinctive aspects of the mechanisms governing OXTR desensitization and internalization in neurons compared to prior studies in heterologous cells.
Collapse
Affiliation(s)
- Kiran George
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hanh T.M. Hoang
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Taryn Tibbs
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Raghavendra Y. Nagaraja
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Guangpu Li
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Eva Troyano-Rodriguez
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mohiuddin Ahmad
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
8
|
Banazadeh M, Abiri A, Poortaheri MM, Asnaashari L, Langarizadeh MA, Forootanfar H. Unexplored power of CRISPR-Cas9 in neuroscience, a multi-OMICs review. Int J Biol Macromol 2024; 263:130413. [PMID: 38408576 DOI: 10.1016/j.ijbiomac.2024.130413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/27/2023] [Accepted: 02/21/2024] [Indexed: 02/28/2024]
Abstract
The neuroscience and neurobiology of gene editing to enhance learning and memory is of paramount interest to the scientific community. The advancements of CRISPR system have created avenues to treat neurological disorders by means of versatile modalities varying from expression to suppression of genes and proteins. Neurodegenerative disorders have also been attributed to non-canonical DNA secondary structures by affecting neuron activity through controlling gene expression, nucleosome shape, transcription, translation, replication, and recombination. Changing DNA regulatory elements which could contribute to the fate and function of neurons are thoroughly discussed in this review. This study presents the ability of CRISPR system to boost learning power and memory, treat or cure genetically-based neurological disorders, and alleviate psychiatric diseases by altering the activity and the irritability of the neurons at the synaptic cleft through DNA manipulation, and also, epigenetic modifications using Cas9. We explore and examine how each different OMIC techniques can come useful when altering DNA sequences. Such insight into the underlying relationship between OMICs and cellular behaviors leads us to better neurological and psychiatric therapeutics by intelligently designing and utilizing the CRISPR/Cas9 technology.
Collapse
Affiliation(s)
- Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ardavan Abiri
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, CT 06520, USA
| | | | - Lida Asnaashari
- Student Research Committee, Kerman Universiy of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Langarizadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Forootanfar
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
9
|
Connor SA, Siddiqui TJ. Synapse organizers as molecular codes for synaptic plasticity. Trends Neurosci 2023; 46:971-985. [PMID: 37652840 DOI: 10.1016/j.tins.2023.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/13/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023]
Abstract
Synapse organizing proteins are multifaceted molecules that coordinate the complex processes of brain development and plasticity at the level of individual synapses. Their importance is demonstrated by the major brain disorders that emerge when their function is compromised. The mechanisms whereby the various families of organizers govern synapses are diverse, but converge on the structure, function, and plasticity of synapses. Therefore, synapse organizers regulate how synapses adapt to ongoing activity, a process central for determining the developmental trajectory of the brain and critical to all forms of cognition. Here, we explore how synapse organizers set the conditions for synaptic plasticity and the associated molecular events, which eventually link to behavioral features of neurodevelopmental and neuropsychiatric disorders. We also propose central questions on how synapse organizers influence network function through integrating nanoscale and circuit-level organization of the brain.
Collapse
Affiliation(s)
- Steven A Connor
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - Tabrez J Siddiqui
- PrairieNeuro Research Centre, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB R3E 0Z3, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; The Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Program in Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
10
|
Zhang R, Jiang H, Liu Y, He G. Structure, function, and pathology of Neurexin-3. Genes Dis 2023; 10:1908-1919. [PMID: 37492720 PMCID: PMC10363586 DOI: 10.1016/j.gendis.2022.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 11/22/2022] Open
Abstract
Neurexin-3 is primarily localized in the presynaptic membrane and forms complexes with various ligands located in the postsynaptic membrane. Neurexin-3 has important roles in synapse development and synapse functions. Neurexin-3 mediates excitatory presynaptic differentiation by interacting with leucine-rich-repeat transmembrane neuronal proteins. Meanwhile, neurexin-3 modulates the expression of presynaptic α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors and γ-aminobutyric acid A receptors by interacting with neuroligins at excitatory and inhibitory synapses. Numerous studies have documented the potential contribution of neurexin-3 to neurodegenerative and neuropsychiatric disorders, such as Alzheimer's disease, addiction behaviors, and other diseases, which raises hopes that understanding the mechanisms of neurexin-3 may hold the key to developing new strategies for related illnesses. This review comprehensively covers the literature to provide current knowledge of the structure, function, and clinical role of neurexin-3.
Collapse
Affiliation(s)
- Rui Zhang
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China
| | - HanXiao Jiang
- Department of Neurology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - YuanJie Liu
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| | - GuiQiong He
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China
- Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
11
|
Pramio DT, Vieceli FM, Varella-Branco E, Goes CP, Kobayashi GS, da Silva Pelegrina DV, de Moraes BC, El Allam A, De Kumar B, Jara G, Farfel JM, Bennett DA, Kundu S, Viapiano MS, Reis EM, de Oliveira PSL, Dos Santos E Passos-Bueno MR, Rothlin CV, Ghosh S, Schechtman D. DNA methylation of the promoter region at the CREB1 binding site is a mechanism for the epigenetic regulation of brain-specific PKMζ. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194909. [PMID: 36682583 PMCID: PMC10037092 DOI: 10.1016/j.bbagrm.2023.194909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/05/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023]
Abstract
Protein kinase M zeta, PKMζ, is a brain enriched kinase with a well characterized role in Long-Term Potentiation (LTP), the activity-dependent strengthening of synapses involved in long-term memory formation. However, little is known about the molecular mechanisms that maintain the tissue specificity of this kinase. Here, we characterized the epigenetic factors, mainly DNA methylation, regulating PKMζ expression in the human brain. The PRKCZ gene has an upstream promoter regulating Protein kinase C ζ (PKCζ), and an internal promoter driving PKMζ expression. A demethylated region, including a canonical CREB binding site, situated at the internal promoter was only observed in human CNS tissues. The induction of site-specific hypermethylation of this region resulted in decreased CREB1 binding and downregulation of PKMζ expression. Noteworthy, CREB binding sites were absent in the upstream promoter of PRKCZ locus, suggesting a specific mechanism for regulating PKMζ expression. These observations were validated using a system of human neuronal differentiation from induced pluripotent stem cells (iPSCs). CREB1 binding at the internal promoter was detected only in differentiated neurons, where PKMζ is expressed. The same epigenetic mechanism in the context of CREB binding site was identified in other genes involved in neuronal differentiation and LTP. Additionally, aberrant DNA hypermethylation at the internal promoter was observed in cases of Alzheimer's disease, correlating with decreased expression of PKMζ in patient brains. Altogether, we present a conserved epigenetic mechanism regulating PKMζ expression and other genes enhanced in the CNS with possible implications in neuronal differentiation and Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | - Carolina Purcell Goes
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil; Laboratory of Neuromodulation of Experimental Pain, Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, SP, Brazil
| | | | | | | | - Aicha El Allam
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | | | - Gabriel Jara
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian National Biosciences Laboratory (LNBio) Campinas, SP, Brazil
| | - José Marcelo Farfel
- Traumatology and Orthopedy Department, Faculdade de Medicina, Universidade de São Paulo, SP, Brazil; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Health Sciences Program, Instituto de Assistência Medica ao Servidor Público do Estado (IAMSPE), SP, Brazil
| | - David Alan Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Somanath Kundu
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Mariano S Viapiano
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Eduardo Moraes Reis
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil
| | - Paulo Sergio Lopes de Oliveira
- Brazilian Center for Research in Energy and Materials (CNPEM), Brazilian National Biosciences Laboratory (LNBio) Campinas, SP, Brazil
| | | | - Carla V Rothlin
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA; Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Sourav Ghosh
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA; Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA.
| | - Deborah Schechtman
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil.
| |
Collapse
|
12
|
Dhume SH, Connor SA, Mills F, Tari PK, Au-Yeung SHM, Karimi B, Oku S, Roppongi RT, Kawabe H, Bamji SX, Wang YT, Brose N, Jackson MF, Craig AM, Siddiqui TJ. Distinct but overlapping roles of LRRTM1 and LRRTM2 in developing and mature hippocampal circuits. eLife 2022; 11:64742. [PMID: 35662394 PMCID: PMC9170246 DOI: 10.7554/elife.64742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 05/20/2022] [Indexed: 01/21/2023] Open
Abstract
LRRTMs are postsynaptic cell adhesion proteins that have region-restricted expression in the brain. To determine their role in the molecular organization of synapses in vivo, we studied synapse development and plasticity in hippocampal neuronal circuits in mice lacking both Lrrtm1 and Lrrtm2. We found that LRRTM1 and LRRTM2 regulate the density and morphological integrity of excitatory synapses on CA1 pyramidal neurons in the developing brain but are not essential for these roles in the mature circuit. Further, they are required for long-term-potentiation in the CA3-CA1 pathway and the dentate gyrus, and for enduring fear memory in both the developing and mature brain. Our data show that LRRTM1 and LRRTM2 regulate synapse development and function in a cell-type and developmental-stage-specific manner, and thereby contribute to the fine-tuning of hippocampal circuit connectivity and plasticity.
Collapse
Affiliation(s)
- Shreya H Dhume
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Steven A Connor
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada.,Department of Biology, York University, Toronto, Canada
| | - Fergil Mills
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Parisa Karimi Tari
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada.,Department of Biology, York University, Toronto, Canada
| | - Sarah H M Au-Yeung
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Benjamin Karimi
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Shinichiro Oku
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Reiko T Roppongi
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Hiroshi Kawabe
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.,Department of Gerontology, Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan.,Department of Pharmacology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Shernaz X Bamji
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Yu Tian Wang
- Division of Neurology, Department of Medicine and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Michael F Jackson
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada
| | - Ann Marie Craig
- Department of Psychiatry and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Tabrez J Siddiqui
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,The Children's Hospital Research Institute of Manitoba, Winnipeg, Canada.,Program in Biomedical Engineering, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
13
|
Transsynaptic cerebellin 4-neogenin 1 signaling mediates LTP in the mouse dentate gyrus. Proc Natl Acad Sci U S A 2022; 119:e2123421119. [PMID: 35544694 DOI: 10.1073/pnas.2123421119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
SignificanceSynapses are controlled by transsynaptic adhesion complexes that mediate bidirectional signaling between pre- and postsynaptic compartments. Long-term potentiation (LTP) of synaptic transmission is thought to enable synaptic modifications during memory formation, but the signaling mechanisms involved remain poorly understood. We show that binding of cerebellin-4 (Cbln4), a secreted ligand of presynaptic neurexin adhesion molecules, to neogenin-1, a postsynaptic surface protein known as a developmental netrin receptor, is essential for normal LTP at entorhinal cortex→dentate gyrus synapses in mice. Cbln4 and neogenin-1 are dispensable for basal synaptic transmission and not involved in establishing synaptic connections as such. Our data identify a netrin receptor as a postsynaptic organizer of synaptic plasticity that collaborates specifically with the presynaptic neurexin-ligand Cbln4.
Collapse
|
14
|
Teng Z, Gottmann K. Hemisynapse Formation Between Target Astrocytes and Cortical Neuron Axons in vitro. Front Mol Neurosci 2022; 15:829506. [PMID: 35386271 PMCID: PMC8978633 DOI: 10.3389/fnmol.2022.829506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 02/08/2022] [Indexed: 01/28/2023] Open
Abstract
One of the most fundamental organizing principles in the mammalian brain is that neurons do not establish synapses with the other major cell type, the astrocytes. However, induced synapse formation between neurons and astrocytes appears conceivable, because astrocytes are well known to express functional ionotropic glutamate receptors. Here, we attempted to trigger synapse formation between co-cultured neurons and astrocytes by overexpressing the strongly synaptogenic adhesion protein LRRTM2 in astrocytes physically contacted by cortical axons. Interestingly, control experiments with immature cortical astrocytes without any overexpression resulted in the induction of synaptic vesicle clustering in contacting axons (hemisynapse formation). This synaptogenic activity correlated with the endogenous expression of the synaptogenic protein Neuroligin1. Hemisynapse formation was further enhanced upon overexpression of LRRTM2 in cortical astrocytes. In contrast, cerebellar astrocytes required overexpression of LRRTM2 for induction of synaptic vesicle clustering in contacting axons. We further addressed, whether hemisynapse formation was accompanied by the appearance of fully functional glutamatergic synapses. We therefore attempted to record AMPA receptor-mediated miniature excitatory postsynaptic currents (mEPSCs) in innervated astrocytes using the whole-cell patch-clamp technique. Despite the endogenous expression of the AMPA receptor subunits GluA2 and to a lesser extent GluA1, we did not reliably observe spontaneous AMPA mEPSCs. In conclusion, overexpression of the synaptogenic protein LRRTM2 induced hemisynapse formation between co-cultured neurons and astrocytes. However, the formation of fully functional synapses appeared to require additional factors critical for nano-alignment of presynaptic vesicles and postsynaptic receptors.
Collapse
|
15
|
Kim J, Park D, Seo NY, Yoon TH, Kim GH, Lee SH, Seo J, Um JW, Lee KJ, Ko J. LRRTM3 regulates activity-dependent synchronization of synapse properties in topographically connected hippocampal neural circuits. Proc Natl Acad Sci U S A 2022; 119:e2110196119. [PMID: 35022233 PMCID: PMC8784129 DOI: 10.1073/pnas.2110196119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 12/03/2021] [Indexed: 11/18/2022] Open
Abstract
Synaptic cell-adhesion molecules (CAMs) organize the architecture and properties of neural circuits. However, whether synaptic CAMs are involved in activity-dependent remodeling of specific neural circuits is incompletely understood. Leucine-rich repeat transmembrane protein 3 (LRRTM3) is required for the excitatory synapse development of hippocampal dentate gyrus (DG) granule neurons. Here, we report that Lrrtm3-deficient mice exhibit selective reductions in excitatory synapse density and synaptic strength in projections involving the medial entorhinal cortex (MEC) and DG granule neurons, accompanied by increased neurotransmitter release and decreased excitability of granule neurons. LRRTM3 deletion significantly reduced excitatory synaptic innervation of hippocampal mossy fibers (Mf) of DG granule neurons onto thorny excrescences in hippocampal CA3 neurons. Moreover, LRRTM3 loss in DG neurons significantly decreased mossy fiber long-term potentiation (Mf-LTP). Remarkably, silencing MEC-DG circuits protected against the decrease in the excitatory synaptic inputs onto DG and CA3 neurons, excitability of DG granule neurons, and Mf-LTP in Lrrtm3-deficient mice. These results suggest that LRRTM3 may be a critical factor in activity-dependent synchronization of the topography of MEC-DG-CA3 excitatory synaptic connections. Collectively, our data propose that LRRTM3 shapes the target-specific structural and functional properties of specific hippocampal circuits.
Collapse
Affiliation(s)
- Jinhu Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Dongseok Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Na-Young Seo
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Neural Circuits Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Taek-Han Yoon
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Gyu Hyun Kim
- Neural Circuits Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Sang-Hoon Lee
- Neural Circuits Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
- Brain Research Core Facilities, KBRI, Daegu 41062, Korea
| | - Jinsoo Seo
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Kea Joo Lee
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
- Neural Circuits Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
| |
Collapse
|
16
|
Liouta K, Chabbert J, Benquet S, Tessier B, Studer V, Sainlos M, De Wit J, Thoumine O, Chamma I. Role of regulatory C-terminal motifs in synaptic confinement of LRRTM2. Biol Cell 2021; 113:492-506. [PMID: 34498765 DOI: 10.1111/boc.202100026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/25/2022]
Abstract
Leucine Rich Repeat Transmembrane proteins (LRRTMs) are neuronal cell adhesion molecules involved in synapse development and plasticity. LRRTM2 is the most synaptogenic isoform of the family, and its expression is strongly restricted to excitatory synapses in mature neurons. However, the mechanisms by which LRRTM2 is trafficked and stabilized at synapses remain unknown. Here, we examine the role of LRRTM2 intracellular domain on its membrane expression and stabilization at excitatory synapses, using a knock-down strategy combined to single molecule tracking and super-resolution dSTORM microscopy. We show that LRRTM2 operates an important shift in mobility after synaptogenesis in hippocampal neurons. Knock-down of LRRTM2 during synapse formation reduced excitatory synapse density in mature neurons. Deletion of LRRTM2 C-terminal domain abolished the compartmentalization of LRRTM2 in dendrites and disrupted its synaptic enrichment. Furtheremore, we show that LRRTM2 diffusion is increased in the absence of its intracellular domain, and that the protein is more dispersed at synapses. Surprisingly, LRRTM2 confinement at synapses was strongly dependent on a YxxC motif in the C-terminal domain, but was independent of the PDZ-like binding motif ECEV. Finally, the nanoscale organization of LRRTM2 at excitatory synapses depended on its C-terminal domain, with involvement of both the PDZ-binding and YxxC motifs. Altogether, these results demonstrate that LRRTM2 trafficking and enrichment at excitatory synapses are dependent on its intracellular domain.
Collapse
Affiliation(s)
- Konstantina Liouta
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Julia Chabbert
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Sebastien Benquet
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Béatrice Tessier
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Vincent Studer
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Matthieu Sainlos
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Joris De Wit
- VIB Center for Brain & Disease Research, Leuven, Belgium.,KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Olivier Thoumine
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Ingrid Chamma
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| |
Collapse
|
17
|
Drulis-Fajdasz D, Gostomska-Pampuch K, Duda P, Wiśniewski JR, Rakus D. Quantitative Proteomics Reveals Significant Differences between Mouse Brain Formations in Expression of Proteins Involved in Neuronal Plasticity during Aging. Cells 2021; 10:2021. [PMID: 34440790 PMCID: PMC8393337 DOI: 10.3390/cells10082021] [Citation(s) in RCA: 8] [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: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022] Open
Abstract
Aging is associated with a general decline in cognitive functions, which appears to be due to alterations in the amounts of proteins involved in the regulation of synaptic plasticity. Here, we present a quantitative analysis of proteins involved in neurotransmission in three brain regions, namely, the hippocampus, the cerebral cortex and the cerebellum, in mice aged 1 and 22 months, using the total protein approach technique. We demonstrate that although the titer of some proteins involved in neurotransmission and synaptic plasticity is affected by aging in a similar manner in all the studied brain formations, in fact, each of the formations represents its own mode of aging. Generally, the hippocampal and cortical proteomes are much more unstable during the lifetime than the cerebellar proteome. The data presented here provide a general picture of the effect of physiological aging on synaptic plasticity and might suggest potential drug targets for anti-aging therapies.
Collapse
Affiliation(s)
- Dominika Drulis-Fajdasz
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| | - Kinga Gostomska-Pampuch
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; (K.G.-P.); (J.R.W.)
- Department of Biochemistry and Immunochemistry, Wrocław Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland
| | - Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| | - Jacek Roman Wiśniewski
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; (K.G.-P.); (J.R.W.)
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| |
Collapse
|
18
|
Ramsey AM, Tang AH, LeGates TA, Gou XZ, Carbone BE, Thompson SM, Biederer T, Blanpied TA. Subsynaptic positioning of AMPARs by LRRTM2 controls synaptic strength. SCIENCE ADVANCES 2021; 7:7/34/eabf3126. [PMID: 34417170 PMCID: PMC8378824 DOI: 10.1126/sciadv.abf3126] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 06/30/2021] [Indexed: 05/07/2023]
Abstract
Recent evidence suggests that nano-organization of proteins within synapses may control the strength of communication between neurons in the brain. The unique subsynaptic distribution of glutamate receptors, which cluster in nanoalignment with presynaptic sites of glutamate release, supports this hypothesis. However, testing it has been difficult because mechanisms controlling subsynaptic organization remain unknown. Reasoning that transcellular interactions could position AMPA receptors (AMPARs), we targeted a key transsynaptic adhesion molecule implicated in controlling AMPAR number, LRRTM2, using engineered, rapid proteolysis. Severing the LRRTM2 extracellular domain led quickly to nanoscale declustering of AMPARs away from release sites, not prompting their escape from synapses until much later. This rapid remodeling of AMPAR position produced significant deficits in evoked, but not spontaneous, postsynaptic receptor activation. These results dissociate receptor numbers from their nanopositioning in determination of synaptic function and support the novel concept that adhesion molecules acutely position receptors to dynamically control synaptic strength.
Collapse
Affiliation(s)
- Austin M Ramsey
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ai-Hui Tang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Tara A LeGates
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Beatrice E Carbone
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Scott M Thompson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
19
|
Matthews PM, Pinggera A, Kampjut D, Greger IH. Biology of AMPA receptor interacting proteins - From biogenesis to synaptic plasticity. Neuropharmacology 2021; 197:108709. [PMID: 34271020 DOI: 10.1016/j.neuropharm.2021.108709] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/19/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022]
Abstract
AMPA-type glutamate receptors mediate the majority of excitatory synaptic transmission in the central nervous system. Their signaling properties and abundance at synapses are both crucial determinants of synapse efficacy and plasticity, and are therefore under sophisticated control. Unique to this ionotropic glutamate receptor (iGluR) is the abundance of interacting proteins that contribute to its complex regulation. These include transient interactions with the receptor cytoplasmic tail as well as the N-terminal domain locating to the synaptic cleft, both of which are involved in AMPAR trafficking and receptor stabilization at the synapse. Moreover, an array of transmembrane proteins operate as auxiliary subunits that in addition to receptor trafficking and stabilization also substantially impact AMPAR gating and pharmacology. Here, we provide an overview of the catalogue of AMPAR interacting proteins, and how they contribute to the complex biology of this central glutamate receptor.
Collapse
Affiliation(s)
- Peter M Matthews
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Alexandra Pinggera
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Domen Kampjut
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
20
|
Abstract
This study presents evidence that the MAGUK family of synaptic scaffolding proteins plays an essential, but redundant, role in long-term potentiation (LTP). The action of PSD-95, but not that of SAP102, requires the binding to the transsynaptic adhesion protein ADAM22, which is required for nanocolumn stabilization. Based on these and previous results, we propose a two-step process in the recruitment of AMPARs during LTP. First, AMPARs, via TARPs, bind to exposed PSD-95 in the PSD. This alone is not adequate to enhance synaptic transmission. Second, the AMPAR/TARP/PSD-95 complex is stabilized in the nanocolumn by binding to ADAM22. A second, ADAM22-independent pathway is proposed for SAP102.
Collapse
|
21
|
Jiang CH, Wei M, Zhang C, Shi YS. The amino-terminal domain of GluA1 mediates LTP maintenance via interaction with neuroplastin-65. Proc Natl Acad Sci U S A 2021; 118:e2019194118. [PMID: 33627404 PMCID: PMC7936340 DOI: 10.1073/pnas.2019194118] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Long-term potentiation (LTP) has long been considered as an important cellular mechanism for learning and memory. LTP expression involves NMDA receptor-dependent synaptic insertion of AMPA receptors (AMPARs). However, how AMPARs are recruited and anchored at the postsynaptic membrane during LTP remains largely unknown. In this study, using CRISPR/Cas9 to delete the endogenous AMPARs and replace them with the mutant forms in single neurons, we have found that the amino-terminal domain (ATD) of GluA1 is required for LTP maintenance. Moreover, we show that GluA1 ATD directly interacts with the cell adhesion molecule neuroplastin-65 (Np65). Neurons lacking Np65 exhibit severely impaired LTP maintenance, and Np65 deletion prevents GluA1 from rescuing LTP in AMPARs-deleted neurons. Thus, our study reveals an essential role for GluA1/Np65 binding in anchoring AMPARs at the postsynaptic membrane during LTP.
Collapse
Affiliation(s)
- Chao-Hua Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing University, 210032 Nanjing, China
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 210032 Nanjing, China
| | - Mengping Wei
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China;
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing University, 210032 Nanjing, China;
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 210032 Nanjing, China
- Institute for Brain Sciences, Nanjing University, 210032 Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, 210032 Nanjing, China
| |
Collapse
|
22
|
Abstract
The function of neuronal circuits relies on the properties of individual neuronal cells and their synapses. We propose that a substantial degree of synapse formation and function is instructed by molecular codes resulting from transcriptional programmes. Recent studies on the Neurexin protein family and its ligands provide fundamental insight into how synapses are assembled and remodelled, how synaptic properties are specified and how single gene mutations associated with neurodevelopmental and psychiatric disorders might modify the operation of neuronal circuits and behaviour. In this Review, we first summarize insights into Neurexin function obtained from various model organisms. We then discuss the mechanisms and logic of the cell type-specific regulation of Neurexin isoforms, in particular at the level of alternative mRNA splicing. Finally, we propose a conceptual framework for how combinations of synaptic protein isoforms act as 'senders' and 'readers' to instruct synapse formation and the acquisition of cell type-specific and synapse-specific functional properties.
Collapse
|
23
|
Kamimura K, Maeda N. Glypicans and Heparan Sulfate in Synaptic Development, Neural Plasticity, and Neurological Disorders. Front Neural Circuits 2021; 15:595596. [PMID: 33679334 PMCID: PMC7928303 DOI: 10.3389/fncir.2021.595596] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are components of the cell surface and extracellular matrix, which bear long polysaccharides called heparan sulfate (HS) attached to the core proteins. HSPGs interact with a variety of ligand proteins through the HS chains, and mutations in HSPG-related genes influence many biological processes and cause various diseases. In particular, recent findings from vertebrate and invertebrate studies have raised the importance of glycosylphosphatidylinositol-anchored HSPGs, glypicans, as central players in the development and functions of synapses. Glypicans are important components of the synapse-organizing protein complexes and serve as ligands for leucine-rich repeat transmembrane neuronal proteins (LRRTMs), leukocyte common antigen-related (LAR) family receptor protein tyrosine phosphatases (RPTPs), and G-protein-coupled receptor 158 (GPR158), regulating synapse formation. Many of these interactions are mediated by the HS chains of glypicans. Neurexins (Nrxs) are also synthesized as HSPGs and bind to some ligands in common with glypicans through HS chains. Therefore, glypicans and Nrxs may act competitively at the synapses. Furthermore, glypicans regulate the postsynaptic expression levels of ionotropic glutamate receptors, controlling the electrophysiological properties and non-canonical BMP signaling of synapses. Dysfunctions of glypicans lead to failures in neuronal network formation, malfunction of synapses, and abnormal behaviors that are characteristic of neurodevelopmental disorders. Recent human genetics revealed that glypicans and HS are associated with autism spectrum disorder, neuroticism, and schizophrenia. In this review, we introduce the studies showing the roles of glypicans and HS in synapse formation, neural plasticity, and neurological disorders, especially focusing on the mouse and Drosophila as potential models for human diseases.
Collapse
Affiliation(s)
- Keisuke Kamimura
- Developmental Neuroscience Project, Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Setagaya, Japan
| | - Nobuaki Maeda
- Developmental Neuroscience Project, Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, Setagaya, Japan
| |
Collapse
|
24
|
Kim HY, Um JW, Ko J. Proper synaptic adhesion signaling in the control of neural circuit architecture and brain function. Prog Neurobiol 2021; 200:101983. [PMID: 33422662 DOI: 10.1016/j.pneurobio.2020.101983] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/23/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Trans-synaptic cell-adhesion molecules are critical for governing various stages of synapse development and specifying neural circuit properties via the formation of multifarious signaling pathways. Recent studies have pinpointed the putative roles of trans-synaptic cell-adhesion molecules in mediating various cognitive functions. Here, we review the literature on the roles of a diverse group of central synaptic organizers, including neurexins (Nrxns), leukocyte common antigen-related receptor protein tyrosine phosphatases (LAR-RPTPs), and their associated binding proteins, in regulating properties of specific type of synapses and neural circuits. In addition, we highlight the findings that aberrant synaptic adhesion signaling leads to alterations in the structures, transmission, and plasticity of specific synapses across diverse brain areas. These results seem to suggest that proper trans-synaptic signaling pathways by Nrxns, LAR-RPTPs, and their interacting network is likely to constitute central molecular complexes that form the basis for cognitive functions, and that these complexes are heterogeneously and complexly disrupted in many neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hee Young Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea; Core Protein Resources Center, DGIST, Daegu, 42988, South Korea.
| | - Jaewon Ko
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, South Korea.
| |
Collapse
|
25
|
Zhang L, Li J, Li Y, Wang Z, Wang G, Yu Y, Song C, Cui W. Spinal caspase-3 contributes to tibial fracture-associated postoperative allodynia via up-regulation of LRRTM1 expression in mice. Neurosci Lett 2020; 739:135429. [PMID: 33069813 DOI: 10.1016/j.neulet.2020.135429] [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: 08/24/2020] [Revised: 09/22/2020] [Accepted: 09/27/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Bone fracture may subsequently cause chronic postoperative pain after orthopedic surgery, but mechanisms remain elusive. The necessity of caspase-3 in neuroinflammation and synaptic plasticity has been summarized in pathological pain. Leucine-rich repeat transmembrane protein 1 (LRRTM1) mediates synaptic delivery of AMPA receptor and synaptogenesis. This study evaluated whether caspase-3 and LRRTM1 are required for fracture-associated postoperative allodynia. METHODS A model of tibial fracture with intramedullary pinning in mice was established for the induction of postoperative pain, verified by measurement of mechanical paw withdrawal threshold and cold scores response to acetone. The caspase-3 specific inhibitor, recombinant caspase-3 and LRRTM1 knockdown by shRNA were utilized for the investigation of pathogenesis as well as the prevention of allodynia. Also, the activity of caspase-3 and the expression of LRRTM1 in the spinal dorsal horn were examined by Western blot and RT-qPCR. RESULTS This study reported that tibial fracture and orthopedic surgery produced long-lasting mechanical allodynia and cold allodynia, along with the up-modulation of spinal caspase-3 activity (but not caspase-3 expression) and LRRTM1 expression. Spinal caspase-3 inhibition prevented fracture-associated behavioral allodynia in a dose-dependent manner. Caspase-3 inhibitor also reduced the spinal increased LRRTM1 level after tibial fracture with pinning. Spinal LRRTM1 deficiency impaired fracture-caused postoperative pain. Intrathecal recombinant caspase-3 facilitated acute pain hypersensitivity and spinal LRRTM1 expression in naïve mice, reversing by LRRTM1 knockdown. CONCLUSION Our current results demonstrate the spinal up-regulation of LRRTM1 by caspase-3 activation in the development of tibial fracture-associated postoperative pain in mice.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Jing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Zhen Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Chengcheng Song
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| | - Wei Cui
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| |
Collapse
|
26
|
Keable R, Leshchyns'ka I, Sytnyk V. Trafficking and Activity of Glutamate and GABA Receptors: Regulation by Cell Adhesion Molecules. Neuroscientist 2020; 26:415-437. [PMID: 32449484 DOI: 10.1177/1073858420921117] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The efficient targeting of ionotropic receptors to postsynaptic sites is essential for the function of chemical excitatory and inhibitory synapses, constituting the majority of synapses in the brain. A growing body of evidence indicates that cell adhesion molecules (CAMs), which accumulate at synapses at the earliest stages of synaptogenesis, are critical for this process. A diverse variety of CAMs assemble into complexes with glutamate and GABA receptors and regulate the targeting of these receptors to the cell surface and synapses. Presynaptically localized CAMs provide an additional level of regulation, sending a trans-synaptic signal that can regulate synaptic strength at the level of receptor trafficking. Apart from controlling the numbers of receptors present at postsynaptic sites, CAMs can also influence synaptic strength by modulating the conductivity of single receptor channels. CAMs thus act to maintain basal synaptic transmission and are essential for many forms of activity dependent synaptic plasticity. These activities of CAMs may underlie the association between CAM gene mutations and synaptic pathology and represent fundamental mechanisms by which synaptic strength is dynamically tuned at both excitatory and inhibitory synapses.
Collapse
Affiliation(s)
- Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
27
|
Neuronal ensemble-specific DNA methylation strengthens engram stability. Nat Commun 2020; 11:639. [PMID: 32005851 PMCID: PMC6994722 DOI: 10.1038/s41467-020-14498-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022] Open
Abstract
Memories are encoded by memory traces or engrams, represented within subsets of neurons that are synchronously activated during learning. However, the molecular mechanisms that drive engram stabilization during consolidation and consequently ensure its reactivation by memory recall are not fully understood. In this study we manipulate, during memory consolidation, the levels of the de novo DNA methyltransferase 3a2 (Dnmt3a2) selectively within dentate gyrus neurons activated by fear conditioning. We found that Dnmt3a2 upregulation enhances memory performance in mice and improves the fidelity of reconstitution of the original neuronal ensemble upon memory retrieval. Moreover, similar manipulation in a sparse, non-engram subset of neurons does not bias engram allocation or modulate memory strength. We further show that neuronal Dnmt3a2 overexpression changes the DNA methylation profile of synaptic plasticity-related genes. Our data implicates DNA methylation selectively within neuronal ensembles as a mechanism of stabilizing engrams during consolidation that supports successful memory retrieval.
Collapse
|
28
|
Cell-to-Cell Communication in Learning and Memory: From Neuro- and Glio-Transmission to Information Exchange Mediated by Extracellular Vesicles. Int J Mol Sci 2019; 21:ijms21010266. [PMID: 31906013 PMCID: PMC6982255 DOI: 10.3390/ijms21010266] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/14/2019] [Accepted: 12/28/2019] [Indexed: 02/06/2023] Open
Abstract
Most aspects of nervous system development and function rely on the continuous crosstalk between neurons and the variegated universe of non-neuronal cells surrounding them. The most extraordinary property of this cellular community is its ability to undergo adaptive modifications in response to environmental cues originating from inside or outside the body. Such ability, known as neuronal plasticity, allows long-lasting modifications of the strength, composition and efficacy of the connections between neurons, which constitutes the biochemical base for learning and memory. Nerve cells communicate with each other through both wiring (synaptic) and volume transmission of signals. It is by now clear that glial cells, and in particular astrocytes, also play critical roles in both modes by releasing different kinds of molecules (e.g., D-serine secreted by astrocytes). On the other hand, neurons produce factors that can regulate the activity of glial cells, including their ability to release regulatory molecules. In the last fifteen years it has been demonstrated that both neurons and glial cells release extracellular vesicles (EVs) of different kinds, both in physiologic and pathological conditions. Here we discuss the possible involvement of EVs in the events underlying learning and memory, in both physiologic and pathological conditions.
Collapse
|
29
|
Dagar S, Gottmann K. Differential Properties of the Synaptogenic Activities of the Neurexin Ligands Neuroligin1 and LRRTM2. Front Mol Neurosci 2019; 12:269. [PMID: 31780894 PMCID: PMC6856695 DOI: 10.3389/fnmol.2019.00269] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/22/2019] [Indexed: 12/03/2022] Open
Abstract
Synaptic cell adhesion molecules are well established to exhibit synaptogenic activity when overexpressed in target cells, indicating that they are involved in formation and functional maturation of synapses. The postsynaptic adhesion proteins Neuroligin1 and LRRTM2 both induce synaptic vesicle clusters in presynaptic axons in vitro by transsynaptically interacting with neurexins. In neurons, this is accompanied by the induction of glutamatergic, but not GABAergic synapses. Although the synaptogenic activity of Neuroligin1 has been well characterized, the properties of the synaptogenic activities of other synaptic adhesion molecules are largely unknown. In this paper, we now compared characteristics of the synaptogenic activities of Neuroligin1 and LRRTM2 upon overexpression in cultured mouse cortical neurons. Individual cortical neurons were transfected with Neuroligin1 and LRRTM2 expression plasmids, respectively, and synaptic vesicle clustering in contacting axons was examined by immunostaining for the vesicle membrane protein VAMP2. In immature neurons at 6–7 days in vitro (DIV) both Neuroligin1 and LRRTM2 exhibited strong synaptogenic activity. However, upon further neuronal differentiation only LRRTM2 retained significant synaptogenic activity at 12–13 DIV. A similar differential developmental maturation of the synaptogenic activities of Neuroligin1 and LRRTM2 was observed for the induction of glutamatergic synapses, which were detected by co-immunostaining for VGLUT1 and Homer1. Most interestingly, the synaptogenic activity of Neuroligin1 was strongly dependent on the expression and function of the synaptic adhesion molecule N-cadherin in immature neurons. In contrast, the synaptogenic activity of LRRTM2 was independent of N-cadherin expression and function in both immature (6–7 DIV) and more mature neurons (14–15 DIV). Taken together, our results with overexpression in cultured cortical neurons revealed striking differences in the properties of the synaptogenic activities of Neuroligin1 and LRRTM2, although both transsynaptically interact with presynaptic neurexins.
Collapse
Affiliation(s)
- Sushma Dagar
- Institute of Neuro- and Sensory Physiology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Kurt Gottmann
- Institute of Neuro- and Sensory Physiology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
30
|
Bissen D, Foss F, Acker-Palmer A. AMPA receptors and their minions: auxiliary proteins in AMPA receptor trafficking. Cell Mol Life Sci 2019; 76:2133-2169. [PMID: 30937469 PMCID: PMC6502786 DOI: 10.1007/s00018-019-03068-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/12/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022]
Abstract
To correctly transfer information, neuronal networks need to continuously adjust their synaptic strength to extrinsic stimuli. This ability, termed synaptic plasticity, is at the heart of their function and is, thus, tightly regulated. In glutamatergic neurons, synaptic strength is controlled by the number and function of AMPA receptors at the postsynapse, which mediate most of the fast excitatory transmission in the central nervous system. Their trafficking to, at, and from the synapse, is, therefore, a key mechanism underlying synaptic plasticity. Intensive research over the last 20 years has revealed the increasing importance of interacting proteins, which accompany AMPA receptors throughout their lifetime and help to refine the temporal and spatial modulation of their trafficking and function. In this review, we discuss the current knowledge about the roles of key partners in regulating AMPA receptor trafficking and focus especially on the movement between the intracellular, extrasynaptic, and synaptic pools. We examine their involvement not only in basal synaptic function, but also in Hebbian and homeostatic plasticity. Included in our review are well-established AMPA receptor interactants such as GRIP1 and PICK1, the classical auxiliary subunits TARP and CNIH, and the newest additions to AMPA receptor native complexes.
Collapse
Affiliation(s)
- Diane Bissen
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany
| | - Franziska Foss
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
31
|
The neurophysiological correlates of handedness: Insights from the lateralized readiness potential. Behav Brain Res 2019; 364:114-122. [DOI: 10.1016/j.bbr.2019.02.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/30/2019] [Accepted: 02/12/2019] [Indexed: 12/21/2022]
|
32
|
Chamma I, Sainlos M, Thoumine O. Biophysical mechanisms underlying the membrane trafficking of synaptic adhesion molecules. Neuropharmacology 2019; 169:107555. [PMID: 30831159 DOI: 10.1016/j.neuropharm.2019.02.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 01/13/2023]
Abstract
Adhesion proteins play crucial roles at synapses, not only by providing a physical trans-synaptic linkage between axonal and dendritic membranes, but also by connecting to functional elements including the pre-synaptic neurotransmitter release machinery and post-synaptic receptors. To mediate these functions, adhesion proteins must be organized on the neuronal surface in a precise and controlled manner. Recent studies have started to describe the mobility, nanoscale organization, and turnover rate of key synaptic adhesion molecules including cadherins, neurexins, neuroligins, SynCAMs, and LRRTMs, and show that some of these proteins are highly mobile in the plasma membrane while others are confined at sub-synaptic compartments, providing evidence for different regulatory pathways. In this review article, we provide a biophysical view of the diffusional trapping of adhesion molecules at synapses, involving both extracellular and intracellular protein interactions. We review the methodology underlying these measurements, including biomimetic systems with purified adhesion proteins, means to perturb protein expression or function, single molecule imaging in cultured neurons, and analytical models to interpret the data. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.
Collapse
Affiliation(s)
- Ingrid Chamma
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France
| | - Matthieu Sainlos
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France
| | - Olivier Thoumine
- Univ. Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France.
| |
Collapse
|
33
|
Hiester BG, Becker MI, Bowen AB, Schwartz SL, Kennedy MJ. Mechanisms and Role of Dendritic Membrane Trafficking for Long-Term Potentiation. Front Cell Neurosci 2018; 12:391. [PMID: 30425622 PMCID: PMC6218485 DOI: 10.3389/fncel.2018.00391] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/11/2018] [Indexed: 01/19/2023] Open
Abstract
Long-term potentiation (LTP) of excitatory synapses is a major form of plasticity for learning and memory in the central nervous system. While the molecular mechanisms of LTP have been debated for decades, there is consensus that LTP induction activates membrane trafficking pathways within dendrites that are essential for synapse growth and strengthening. Current models suggest that key molecules for synaptic potentiation are sequestered within intracellular organelles, which are mobilized by synaptic activity to fuse with the plasma membrane following LTP induction. While the identity of the factors mobilized to the plasma membrane during LTP remain obscure, the field has narrowly focused on AMPA-type glutamate receptors. Here, we review recent literature and present new experimental data from our lab investigating whether AMPA receptors trafficked from intracellular organelles directly contribute to synaptic strengthening during LTP. We propose a modified model where membrane trafficking delivers distinct factors that are required to maintain synapse growth and AMPA receptor incorporation following LTP. Finally, we pose several fundamental questions that may guide further inquiry into the role of membrane trafficking for synaptic plasticity.
Collapse
Affiliation(s)
- Brian G Hiester
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Matthew I Becker
- Department of Physiology and Biophysics, 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
| | - Samantha L Schwartz
- 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
|
34
|
Dynamics, nanoscale organization, and function of synaptic adhesion molecules. Mol Cell Neurosci 2018; 91:95-107. [DOI: 10.1016/j.mcn.2018.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
|
35
|
Schroeder A, Vanderlinden J, Vints K, Ribeiro LF, Vennekens KM, Gounko NV, Wierda KD, de Wit J. A Modular Organization of LRR Protein-Mediated Synaptic Adhesion Defines Synapse Identity. Neuron 2018; 99:329-344.e7. [PMID: 29983322 DOI: 10.1016/j.neuron.2018.06.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/04/2018] [Accepted: 06/14/2018] [Indexed: 10/28/2022]
Abstract
Pyramidal neurons express rich repertoires of leucine-rich repeat (LRR)-containing adhesion molecules with similar synaptogenic activity in culture. The in vivo relevance of this molecular diversity is unclear. We show that hippocampal CA1 pyramidal neurons express multiple synaptogenic LRR proteins that differentially distribute to the major excitatory inputs on their apical dendrites. At Schaffer collateral (SC) inputs, FLRT2, LRRTM1, and Slitrk1 are postsynaptically localized and differentially regulate synaptic structure and function. FLRT2 controls spine density, whereas LRRTM1 and Slitrk1 exert opposing effects on synaptic vesicle distribution at the active zone. All LRR proteins differentially affect synaptic transmission, and their combinatorial loss results in a cumulative phenotype. At temporoammonic (TA) inputs, LRRTM1 is absent; FLRT2 similarly controls functional synapse number, whereas Slitrk1 function diverges to regulate postsynaptic AMPA receptor density. Thus, LRR proteins differentially control synaptic architecture and function and act in input-specific combinations and a context-dependent manner to specify synaptic properties.
Collapse
Affiliation(s)
- Anna Schroeder
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Jeroen Vanderlinden
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Katlijn Vints
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium; Electron Microscopy Platform & VIB BioImaging Core, Herestraat 49, 3000 Leuven, Belgium
| | - Luís F Ribeiro
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Kristel M Vennekens
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Natalia V Gounko
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium; Electron Microscopy Platform & VIB BioImaging Core, Herestraat 49, 3000 Leuven, Belgium
| | - Keimpe D Wierda
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
36
|
Deletion of LRRTM1 and LRRTM2 in adult mice impairs basal AMPA receptor transmission and LTP in hippocampal CA1 pyramidal neurons. Proc Natl Acad Sci U S A 2018; 115:E5382-E5389. [PMID: 29784826 DOI: 10.1073/pnas.1803280115] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Leucine-rich repeat transmembrane (LRRTM) proteins are synaptic cell adhesion molecules that influence synapse formation and function. They are genetically associated with neuropsychiatric disorders, and via their synaptic actions likely regulate the establishment and function of neural circuits in the mammalian brain. Here, we take advantage of the generation of a LRRTM1 and LRRTM2 double conditional knockout mouse (LRRTM1,2 cKO) to examine the role of LRRTM1,2 at mature excitatory synapses in hippocampal CA1 pyramidal neurons. Genetic deletion of LRRTM1,2 in vivo in CA1 neurons using Cre recombinase-expressing lentiviruses dramatically impaired long-term potentiation (LTP), an impairment that was rescued by simultaneous expression of LRRTM2, but not LRRTM4. Mutation or deletion of the intracellular tail of LRRTM2 did not affect its ability to rescue LTP, while point mutations designed to impair its binding to presynaptic neurexins prevented rescue of LTP. In contrast to previous work using shRNA-mediated knockdown of LRRTM1,2, KO of these proteins at mature synapses also caused a decrease in AMPA receptor-mediated, but not NMDA receptor-mediated, synaptic transmission and had no detectable effect on presynaptic function. Imaging of recombinant photoactivatable AMPA receptor subunit GluA1 in the dendritic spines of cultured neurons revealed that it was less stable in the absence of LRRTM1,2. These results illustrate the advantages of conditional genetic deletion experiments for elucidating the function of endogenous synaptic proteins and suggest that LRRTM1,2 proteins help stabilize synaptic AMPA receptors at mature spines during basal synaptic transmission and LTP.
Collapse
|
37
|
Leucine-rich repeat-containing synaptic adhesion molecules as organizers of synaptic specificity and diversity. Exp Mol Med 2018; 50:1-9. [PMID: 29628503 PMCID: PMC5938020 DOI: 10.1038/s12276-017-0023-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022] Open
Abstract
The brain harbors billions of neurons that form distinct neural circuits with exquisite specificity. Specific patterns of connectivity between distinct neuronal cell types permit the transfer and computation of information. The molecular correlates that give rise to synaptic specificity are incompletely understood. Recent studies indicate that cell-surface molecules are important determinants of cell type identity and suggest that these are essential players in the specification of synaptic connectivity. Leucine-rich repeat (LRR)-containing adhesion molecules in particular have emerged as key organizers of excitatory and inhibitory synapses. Here, we discuss emerging evidence that LRR proteins regulate the assembly of specific connectivity patterns across neural circuits, and contribute to the diverse structural and functional properties of synapses, two key features that are critical for the proper formation and function of neural circuits. Further analysis of synaptic proteins will provide insights into the functioning of neural circuits and associated brain disorders. The brain houses numerous highly specialized neuron types, which transfer and process information via a complex network of synaptic connections. Every neuron develops its own distinctive synapses with specific functions, but exactly how this is achieved is not clear. Joris de Wit and Anna Schroeder at the VIB Center for Brain and Disease Research in Leuven, Belgium, reviewed recent research into the leucine-rich repeat-containing (LRR) proteins, which are thought to be major organizers of synaptic connectivity and key regulators of healthy neural circuit development. Further investigations into the functionality of LRR proteins in the brain will not only improve understanding of neural circuitry but also provide insights into synaptic impairments in brain disorders like schizophrenia.
Collapse
|
38
|
Jurado S. AMPA Receptor Trafficking in Natural and Pathological Aging. Front Mol Neurosci 2018; 10:446. [PMID: 29375307 PMCID: PMC5767248 DOI: 10.3389/fnmol.2017.00446] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/21/2017] [Indexed: 01/09/2023] Open
Abstract
α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) enable most excitatory transmission in the brain and are crucial for mediating basal synaptic strength and plasticity. Because of the importance of their function, AMPAR dynamics, activity and subunit composition undergo a tight regulation which begins as early as prenatal development and continues through adulthood. Accumulating evidence suggests that the precise regulatory mechanisms involved in orchestrating AMPAR trafficking are challenged in the aging brain. In turn dysregulation of AMPARs can be linked to most neurological and neurodegenerative disorders. Understanding the mechanisms that govern AMPAR signaling during natural and pathological cognitive decline will guide the efforts to develop most effective ways to tackle neurodegenerative diseases which are one of the primary burdens afflicting an increasingly aging population. In this review, I provide a brief overview of the molecular mechanisms involved in AMPAR trafficking highlighting what is currently known about how these processes change with age and disease. As a particularly well-studied example of AMPAR dysfunction in pathological aging I focus in Alzheimer’s disease (AD) with special emphasis in how the production of neurofibrillary tangles (NFTs) and amyloid-β plaques may contribute to disruption in AMPAR function.
Collapse
Affiliation(s)
- Sandra Jurado
- Instituto de Neurociencias CSIC-UMH, San Juan de Alicante, Spain
| |
Collapse
|
39
|
Abstract
At each of the brain's vast number of synapses, the presynaptic nerve terminal, synaptic cleft, and postsynaptic specialization form a transcellular unit to enable efficient transmission of information between neurons. While we know much about the molecular machinery within each compartment, we are only beginning to understand how these compartments are structurally registered and functionally integrated with one another. This review will describe the organization of each compartment and then discuss their alignment across pre- and postsynaptic cells at a nanometer scale. We propose that this architecture may allow for precise synaptic information exchange and may be modulated to contribute to the remarkable plasticity of brain function.
Collapse
Affiliation(s)
- Thomas Biederer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA.
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
40
|
Biederer T, Kaeser PS, Blanpied TA. Transcellular Nanoalignment of Synaptic Function. Neuron 2017; 96:680-696. [PMID: 29096080 PMCID: PMC5777221 DOI: 10.1016/j.neuron.2017.10.006] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/29/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022]
Abstract
At each of the brain's vast number of synapses, the presynaptic nerve terminal, synaptic cleft, and postsynaptic specialization form a transcellular unit to enable efficient transmission of information between neurons. While we know much about the molecular machinery within each compartment, we are only beginning to understand how these compartments are structurally registered and functionally integrated with one another. This review will describe the organization of each compartment and then discuss their alignment across pre- and postsynaptic cells at a nanometer scale. We propose that this architecture may allow for precise synaptic information exchange and may be modulated to contribute to the remarkable plasticity of brain function.
Collapse
Affiliation(s)
- Thomas Biederer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA.
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
41
|
Jang S, Lee H, Kim E. Synaptic adhesion molecules and excitatory synaptic transmission. Curr Opin Neurobiol 2017; 45:45-50. [DOI: 10.1016/j.conb.2017.03.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/04/2017] [Accepted: 03/15/2017] [Indexed: 10/19/2022]
|
42
|
The Brain-Enriched MicroRNA miR-9-3p Regulates Synaptic Plasticity and Memory. J Neurosci 2017; 36:8641-52. [PMID: 27535911 DOI: 10.1523/jneurosci.0630-16.2016] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 06/28/2016] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED MicroRNAs (miRNAs) are small, noncoding RNAs that posttranscriptionally regulate gene expression in many tissues. Although a number of brain-enriched miRNAs have been identified, only a few specific miRNAs have been revealed as critical regulators of synaptic plasticity, learning, and memory. miR-9-5p/3p are brain-enriched miRNAs known to regulate development and their changes have been implicated in several neurological disorders, yet their role in mature neurons in mice is largely unknown. Here, we report that inhibition of miR-9-3p, but not miR-9-5p, impaired hippocampal long-term potentiation (LTP) without affecting basal synaptic transmission. Moreover, inhibition of miR-9-3p in the hippocampus resulted in learning and memory deficits. Furthermore, miR-9-3p inhibition increased the expression of the LTP-related genes Dmd and SAP97, the expression levels of which are negatively correlated with LTP. These results suggest that miR-9-3p-mediated gene regulation plays important roles in synaptic plasticity and hippocampus-dependent memory. SIGNIFICANCE STATEMENT Despite the abundant expression of the brain-specific microRNA miR-9-5p/3p in both proliferating and postmitotic neurons, most functional studies have focused on their role in neuronal development. Here, we examined the role of miR-9-5p/3p in adult brain and found that miR-9-3p, but not miR-9-5p, has a critical role in hippocampal synaptic plasticity and memory. Moreover, we identified in vivo binding targets of miR-9-3p that are involved in the regulation of long-term potentiation. Our study provides the very first evidence for the critical role of miR-9-3p in synaptic plasticity and memory in the adult mouse.
Collapse
|
43
|
Subunit-specific role for the amino-terminal domain of AMPA receptors in synaptic targeting. Proc Natl Acad Sci U S A 2017. [PMID: 28630296 DOI: 10.1073/pnas.1707472114] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The amino-terminal domain (ATD) of AMPA receptors (AMPARs) accounts for approximately 50% of the protein, yet its functional role, if any, remains a mystery. We have discovered that the translocation of surface GluA1, but not GluA2, AMPAR subunits to the synapse requires the ATD. GluA1A2 heteromers in which the ATD of GluA1 is absent fail to translocate, establishing a critical role of the ATD of GluA1. Inserting GFP into the ATD interferes with the constitutive synaptic trafficking of GluA1, but not GluA2, mimicking the deletion of the ATD. Remarkably, long-term potentiation (LTP) can override the masking effect of the GFP tag. GluA1, but not GluA2, lacking the ATD fails to show LTP. These findings uncover a role for the ATD in subunit-specific synaptic trafficking of AMPARs, both constitutively and during plasticity. How LTP, induced postsynaptically, engages these extracellular trafficking motifs and what specific cleft proteins participate in the process remain to be elucidated.
Collapse
|
44
|
Watson JF, Ho H, Greger IH. Synaptic transmission and plasticity require AMPA receptor anchoring via its N-terminal domain. eLife 2017; 6. [PMID: 28290985 PMCID: PMC5370185 DOI: 10.7554/elife.23024] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 03/04/2017] [Indexed: 12/02/2022] Open
Abstract
AMPA-type glutamate receptors (AMPARs) mediate fast excitatory neurotransmission and are selectively recruited during activity-dependent plasticity to increase synaptic strength. A prerequisite for faithful signal transmission is the positioning and clustering of AMPARs at postsynaptic sites. The mechanisms underlying this positioning have largely been ascribed to the receptor cytoplasmic C-termini and to AMPAR-associated auxiliary subunits, both interacting with the postsynaptic scaffold. Here, using mouse organotypic hippocampal slices, we show that the extracellular AMPAR N-terminal domain (NTD), which projects midway into the synaptic cleft, plays a fundamental role in this process. This highly sequence-diverse domain mediates synaptic anchoring in a subunit-selective manner. Receptors lacking the NTD exhibit increased mobility in synapses, depress synaptic transmission and are unable to sustain long-term potentiation (LTP). Thus, synaptic transmission and the expression of LTP are dependent upon an AMPAR anchoring mechanism that is driven by the NTD. DOI:http://dx.doi.org/10.7554/eLife.23024.001 Neurons send signals via electrical impulses that are transmitted between cells by small molecules known as neurotransmitters. The information is passed from neuron to neuron at specialized points of contact termed synapses. On release of neurotransmitters from the first neuron, the molecules attach to ‘docking stations’ called receptors on the next neuron, referred to as the postsynaptic cell. One of these receptors, the AMPA receptor, transmits signals by binding to a neurotransmitter called glutamate. Previous research has shown that in order to bind glutamate effectively, these receptors need to be trapped and anchored at the correct location at the synapse. This trapping mechanism controls the number of receptors present, which strengthens the synapse, and ultimately mediates learning and memory. However, it is still not clear how AMPA receptor trapping is achieved. To investigate this question, Watson et al. examined how AMPA receptors (and mutant forms of the receptor) affect the communication between neurons using brain slices from mice. The experiments show that an external segment of the AMPA receptor called the N-terminal domain (or NTD for short) is a key element for receptor anchoring at the postsynapse. The AMPA receptor is made out of four different subunits; when the NTD portion was removed from one specific subunit, fewer receptors were anchored correctly at the postsynapse. When the NTD was removed from another subunit, it completely prevented the synapse from learning. Therefore, the NTD brings about subunit-selective anchoring of the AMPA receptor, which affects the ability of the synapse to transmit signals. Important next steps would be to identify the proteins that interact with the NTD and how this specific anchoring affects the strength of the synapse. Another key step will be to understand what mechanisms control the number of AMPA receptors at synapses, to ultimately enable learning. DOI:http://dx.doi.org/10.7554/eLife.23024.002
Collapse
Affiliation(s)
- Jake F Watson
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Hinze Ho
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
45
|
Vadakkan KI. The functional role of all postsynaptic potentials examined from a first-person frame of reference. Rev Neurosci 2016; 27:159-84. [PMID: 26540219 DOI: 10.1515/revneuro-2015-0036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/10/2015] [Indexed: 01/04/2023]
Abstract
When assigning a central role to the neuronal firing, a large number of incoming postsynaptic potentials not utilized during both supra- and subthreshold neuronal activations are not given any functional significance. Local synaptic potentials at the apical dendrites get attenuated as they arrive at the soma to nearly a twentieth of what a synapse proximal to the soma produces. Conservation of these functions necessitates searching for their functional roles. Potentials induced at the postsynapses of neurons of all the neuronal orders activated by sensory inputs carry small bits of sensory information. The activation of these postsynapses by any means other than the activation from their corresponding presynaptic terminals, that also contribute to oscillating potentials, induce the semblance of the arrival of activity from their presynaptic terminals. This is a candidate mechanism for inducing the first-person internal sensory elements of various higher brain functions as a systems property. They also contribute to the firing of subthreshold-activated neurons, including motor neurons. Operational mechanism of inter-postsynaptic functional LINKs can provide necessary structural requirements for these functions. The functional independence of the distal dendritic compartment and recent evidence for in vivo dendritic spikes indicate their independent role in the formation of internal sensory elements. In these contexts, a neuronal soma is flanked by a large number of quasi-functional internal sensory processing units operated using very little energy, even when a neuron is not firing. A large number of possible combinations of internal sensory units explains the corresponding number of specific memory retrievals by the system in response to various cue stimuli.
Collapse
|
46
|
Roppongi RT, Karimi B, Siddiqui TJ. Role of LRRTMs in synapse development and plasticity. Neurosci Res 2016; 116:18-28. [PMID: 27810425 DOI: 10.1016/j.neures.2016.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/10/2016] [Accepted: 10/14/2016] [Indexed: 12/19/2022]
Abstract
Leucine-rich-repeat transmembrane neuronal proteins (LRRTMs) are a family of four synapse organizing proteins critical for the development and function of excitatory synapses. The genes encoding LRRTMs and their binding partners, neurexins and HSPGs, are strongly associated with multiple psychiatric disorders. Here, we review the literature covering their structural features, expression patterns in the developing and adult brains, evolutionary origins, and discovery as synaptogenic proteins. We also discuss their role in the development and plasticity of excitatory synapses as well as their disease associations.
Collapse
Affiliation(s)
- Reiko T Roppongi
- Department of Physiology and Pathophysiology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada; Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, 710 William Avenue, Winnipeg R3Y 0Z3, MB, Canada
| | - Benyamin Karimi
- Department of Physiology and Pathophysiology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada; Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, 710 William Avenue, Winnipeg R3Y 0Z3, MB, Canada
| | - Tabrez J Siddiqui
- Department of Physiology and Pathophysiology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada; Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, 710 William Avenue, Winnipeg R3Y 0Z3, MB, Canada.
| |
Collapse
|
47
|
Comparison of protein repeat classifications based on structure and sequence families. Biochem Soc Trans 2016; 43:832-7. [PMID: 26517890 DOI: 10.1042/bst20150079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tandem repeats (TR) in proteins are common in nature and have several unique functions. They come in various forms that are frequently difficult to recognize from a sequence. A previously proposed structural classification has been recently implemented in the RepeatsDB database. This defines five main classes, mainly based on repeat unit length, with subclasses representing specific folds. Sequence-based classifications, such as Pfam, provide an alternative classification based on evolutionarily conserved repeat families. Here, we discuss a detailed comparison between the structural classes in RepeatsDB and the corresponding Pfam repeat families and clans. Most instances are found to map one-to-one between structure and sequence. Some notable exceptions such as leucine-rich repeats (LRRs) and α-solenoids are discussed.
Collapse
|
48
|
Abstract
AMPA receptors (AMPARs) are assemblies of four core subunits, GluA1-4, that mediate most fast excitatory neurotransmission. The component subunits determine the functional properties of AMPARs, and the prevailing view is that the subunit composition also determines AMPAR trafficking, which is dynamically regulated during development, synaptic plasticity and in response to neuronal stress in disease. Recently, the subunit dependence of AMPAR trafficking has been questioned, leading to a reappraisal of this field. In this Review, we discuss what is known, uncertain, conjectured and unknown about the roles of the individual subunits, and how they affect AMPAR assembly, trafficking and function under both normal and pathological conditions.
Collapse
|
49
|
Kuhn PH, Colombo AV, Schusser B, Dreymueller D, Wetzel S, Schepers U, Herber J, Ludwig A, Kremmer E, Montag D, Müller U, Schweizer M, Saftig P, Bräse S, Lichtenthaler SF. Systematic substrate identification indicates a central role for the metalloprotease ADAM10 in axon targeting and synapse function. eLife 2016; 5. [PMID: 26802628 PMCID: PMC4786429 DOI: 10.7554/elife.12748] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/22/2016] [Indexed: 12/11/2022] Open
Abstract
Metzincin metalloproteases have major roles in intercellular communication by modulating the function of membrane proteins. One of the proteases is the a-disintegrin-and-metalloprotease 10 (ADAM10) which acts as alpha-secretase of the Alzheimer's disease amyloid precursor protein. ADAM10 is also required for neuronal network functions in murine brain, but neuronal ADAM10 substrates are only partly known. With a proteomic analysis of Adam10-deficient neurons we identified 91, mostly novel ADAM10 substrate candidates, making ADAM10 a major protease for membrane proteins in the nervous system. Several novel substrates, including the neuronal cell adhesion protein NrCAM, are involved in brain development. Indeed, we detected mistargeted axons in the olfactory bulb of conditional ADAM10-/- mice, which correlate with reduced cleavage of NrCAM, NCAM and other ADAM10 substrates. In summary, the novel ADAM10 substrates provide a molecular basis for neuronal network dysfunctions in conditional ADAM10-/- mice and demonstrate a fundamental function of ADAM10 in the brain.
Collapse
Affiliation(s)
- Peer-Hendrik Kuhn
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Institut für Pathologie und Pathologische Anatomie, Technische Universität München, Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany
| | - Alessio Vittorio Colombo
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany
| | - Benjamin Schusser
- Department of Animal Science, Institute for Animal Physiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniela Dreymueller
- Institute of Pharmacology and Toxicology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Sebastian Wetzel
- Institute of Biochemistry, Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Ute Schepers
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Julia Herber
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Elisabeth Kremmer
- German Research Center for Environmental Health, Institute of Molecular Tumor immunology, Helmholtz Zentrum München, Munich, Germany
| | - Dirk Montag
- Neurogenetics, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Ulrike Müller
- Department of Functional Genomics, Institute for Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Michaela Schweizer
- Service-Gruppe für Elektronenmikroskopie, Zentrum für Molekulare Neurobiologie, Hamburg, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Stefan Bräse
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Stefan F Lichtenthaler
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
50
|
Um JW, Choi TY, Kang H, Cho YS, Choii G, Uvarov P, Park D, Jeong D, Jeon S, Lee D, Kim H, Lee SH, Bae YC, Choi SY, Airaksinen MS, Ko J. LRRTM3 Regulates Excitatory Synapse Development through Alternative Splicing and Neurexin Binding. Cell Rep 2016; 14:808-822. [PMID: 26776509 DOI: 10.1016/j.celrep.2015.12.081] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/29/2015] [Accepted: 12/16/2015] [Indexed: 11/27/2022] Open
Abstract
The four members of the LRRTM family (LRRTM1-4) are postsynaptic adhesion molecules essential for excitatory synapse development. They have also been implicated in neuropsychiatric diseases. Here, we focus on LRRTM3, showing that two distinct LRRTM3 variants generated by alternative splicing regulate LRRTM3 interaction with PSD-95, but not its excitatory synapse-promoting activity. Overexpression of either LRRTM3 variant increased excitatory synapse density in dentate gyrus (DG) granule neurons, whereas LRRTM3 knockdown decreased it. LRRTM3 also controlled activity-regulated AMPA receptor surface expression in an alternative splicing-dependent manner. Furthermore, Lrrtm3-knockout mice displayed specific alterations in excitatory synapse density, excitatory synaptic transmission and excitability in DG granule neurons but not in CA1 pyramidal neurons. Lastly, LRRTM3 required only specific splice variants of presynaptic neurexins for their synaptogenic activity. Collectively, our data highlight alternative splicing and differential presynaptic ligand utilization in the regulation of LRRTMs, revealing key regulatory mechanisms for excitatory synapse development.
Collapse
Affiliation(s)
- Ji Won Um
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea; Department of Physiology and BK21 PLUS Project to Medical Sciences, Yonsei University College of Medicine, Seoul 120-751, Korea
| | - Tae-Yong Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul 110-749, Korea
| | - Hyeyeon Kang
- Department of Physiology and BK21 PLUS Project to Medical Sciences, Yonsei University College of Medicine, Seoul 120-751, Korea
| | - Yi Sul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Gayoung Choii
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Pavel Uvarov
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki 14, Finland
| | - Dongseok Park
- Department of Physiology and BK21 PLUS Project to Medical Sciences, Yonsei University College of Medicine, Seoul 120-751, Korea
| | - Daun Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Sangmin Jeon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Dongmin Lee
- Department of Anatomy and Neuroscience, Korea 21 Biomedical Science, College of Medicine, Korea University, 126-1, 5-ka, Anam-dong, Seongbuk-gu, Seoul 136-705, Korea
| | - Hyun Kim
- Department of Anatomy and Neuroscience, Korea 21 Biomedical Science, College of Medicine, Korea University, 126-1, 5-ka, Anam-dong, Seongbuk-gu, Seoul 136-705, Korea
| | - Seung-Hee Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Yong-Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul 110-749, Korea
| | - Matti S Airaksinen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki 14, Finland
| | - Jaewon Ko
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea.
| |
Collapse
|