1
|
Qneibi M, Bdir S, Bdair M, Aldwaik SA, Heeh M, Sandouka D, Idais T. Exploring the role of AMPA receptor auxiliary proteins in synaptic functions and diseases. FEBS J 2025; 292:2433-2478. [PMID: 39394632 DOI: 10.1111/febs.17287] [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: 05/18/2024] [Revised: 08/21/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) ionotropic glutamate receptors (AMPARs) mediate rapid excitatory synaptic transmission in the mammalian brain, primarily driven by the neurotransmitter glutamate. The modulation of AMPAR activity, particularly calcium-permeable AMPARs (CP-AMPARs), is crucially influenced by various auxiliary subunits. These subunits are integral membrane proteins that bind to the receptor's core and modify its functional properties, including ion channel kinetics and receptor trafficking. This review comprehensively catalogs all known AMPAR auxiliary proteins, providing vital insights into the biochemical mechanisms governing synaptic modulation and the specific impact of CP-AMPARs compared to their calcium-impermeable AMPA receptor (CI-AMPARs). Understanding the complex interplay between AMPARs and their auxiliary subunits in different brain regions is essential for elucidating their roles in cognitive functions such as learning and memory. Importantly, alterations in these auxiliary proteins' expression, function or interactions have been implicated in various neurological disorders. Aberrant signaling through CP-AMPARs, in particular, is associated with severe synaptic dysfunctions across neurodevelopmental, neurodegenerative and psychiatric conditions. Targeting the distinct properties of AMPAR-auxiliary subunit complexes, especially those involving CP-AMPARs, could disclose new therapeutic strategies, potentially allowing for more precise interventions in treating complex neuronal disorders.
Collapse
Affiliation(s)
- Mohammad Qneibi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Sosana Bdir
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammad Bdair
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Samia Ammar Aldwaik
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | | | - Dana Sandouka
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Tala Idais
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| |
Collapse
|
2
|
Miguez-Cabello F, Wang XT, Yan Y, Brake N, Alexander RPD, Perozzo AM, Khadra A, Bowie D. GluA2-containing AMPA receptors form a continuum of Ca 2+-permeable channels. Nature 2025; 641:537-544. [PMID: 40108453 DOI: 10.1038/s41586-025-08736-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025]
Abstract
Fast excitatory neurotransmission in the mammalian brain is mediated by cation-selective AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors (AMPARs)1. AMPARs are critical for the learning and memory mechanisms of Hebbian plasticity2 and glutamatergic synapse homeostasis3, with recent work establishing that AMPAR missense mutations can cause autism and intellectual disability4-7. AMPARs have been grouped into two functionally distinct tetrameric assemblies based on the inclusion or exclusion of the GluA2 subunit that determines Ca2+ permeability through RNA editing8,9. GluA2-containing AMPARs are the most abundant in the central nervous system and considered to be Ca2+ impermeable10. Here we show this is not the case. Contrary to conventional understanding, GluA2-containing AMPARs form a continuum of polyamine-insensitive ion channels with varying degrees of Ca2+ permeability. Their ability to transport Ca2+ is shaped by the subunit composition of AMPAR tetramers as well as the spatial orientation of transmembrane AMPAR regulatory proteins and cornichon auxiliary subunits. Ca2+ crosses the ion-conduction pathway by docking to an extracellular binding site that helps funnel divalent ions into the pore selectivity filter. The dynamic range in Ca2+ permeability, however, arises because auxiliary subunits primarily modify the selectivity filter. Taken together, our work proposes a broader role for AMPARs in Ca2+ signalling in the mammalian brain and offers mechanistic insight into the pathogenic nature of missense mutations.
Collapse
Affiliation(s)
| | - Xin-Tong Wang
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Yuhao Yan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Niklas Brake
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- Quantitative Life Sciences PhD program, McGill University, Montreal, Quebec, Canada
| | - Ryan P D Alexander
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Amanda M Perozzo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Anmar Khadra
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Derek Bowie
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
3
|
Jing W, Qingqing C, Xia Y, Ningxiang Q, Demei X, Xuefeng W, Ming A, Xi P, Liang W. Inhibiting SNX14 Alleviates Epileptic Seizures by Regulating GluA2 Degradation via the Lysosomal Pathway. Mol Neurobiol 2025:10.1007/s12035-025-04945-y. [PMID: 40237949 DOI: 10.1007/s12035-025-04945-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
Epilepsy is a common chronic neurological condition, and temporal lobe epilepsy (TLE) is the most common form of refractory epilepsy. However, the underlying causes of TLE remain unclear. Our initial findings revealed that the expression of sorting nexin 14 (SNX14), which is a member of the sorting nexin protein family, was significantly upregulated in brain tissues from both patients with TLE and mouse models of TLE. Moreover, modulation of SNX14 expression in the hippocampus of mice demonstrated that SNX14 downregulation significantly decreased the susceptibility to and severity of seizures, whereas SNX14 overexpression exerted the opposite effects. Mechanistically, we revealed that GluA2, which is a subunit of the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor, was a downstream target of SNX14. Further studies indicated that SNX14 modulated GluA2 protein levels by regulating GluA2 degradation via the lysosomal pathway, which in turn influenced glutamatergic synaptic transmission. In conclusion, our findings suggest that the SNX14-GluA2 pathway could be a promising target for the development of novel treatments for epilepsy.
Collapse
Affiliation(s)
- Wang Jing
- Department of Neurology, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Cao Qingqing
- Department of Neurology, Bishan Hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, China
| | - Yan Xia
- Department of Neurology, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Ningxiang
- Department of Neurology, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xu Demei
- Department of Neurology, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wang Xuefeng
- Department of Neurology, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ai Ming
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Xi
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Wang Liang
- Department of Neurology, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Italia M, Spinola A, Borroni B, DiLuca M, Gardoni F. Long-term exposure to anti-GluA3 antibodies triggers functional and structural changes in hippocampal neurons. Neurobiol Dis 2025; 207:106843. [PMID: 39954743 DOI: 10.1016/j.nbd.2025.106843] [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: 10/08/2024] [Revised: 01/15/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025] Open
Abstract
Autoantibodies targeting the GluA3 subunit of AMPA receptors (AMPARs) are implicated in various neurological disorders, including Rasmussen's encephalitis, epilepsy, and frontotemporal dementia. However, their precise role in disease pathology remains insufficiently understood. This study investigated the long-term effects of human anti-GluA3 antibodies (anti-GluA3 hIgGs) on neuronal morphology and function using primary rat hippocampal neurons. We found that long-term exposure to anti-GluA3 hIgGs leads to the delocalisation of GluA3-containing AMPARs at extrasynaptic sites. This molecular event is correlated to dendritic arbor reorganisation, characterised by increased complexity near the soma and progressive simplification in distal regions as well as an increase in the number of shorter dendrites and a corresponding loss of longer ones, thus suggesting altered dendritic pruning dynamics. The altered neuronal architecture was accompanied by an increase in the number of dendritic spines and a modification of their morphology, indicating relevant changes in synaptic connectivity. Functionally, anti-GluA3 hIgGs significantly enhanced NMDA receptor-mediated postsynaptic Ca2+ currents and increased nuclear levels of phosphorylated cAMP response element-binding protein (CREB), indicating altered signal transduction. Overall, our study provides critical insights into the role of anti-GluA3 hIgGs in disease and potentially identifies new therapeutic targets for pathological conditions where they are present.
Collapse
Affiliation(s)
- Maria Italia
- Department of Pharmacological and Biomolecular Sciences (DiSFeB) "Rodolfo Paoletti", University of Milan, Milan, Italy
| | - Alessio Spinola
- Department of Pharmacological and Biomolecular Sciences (DiSFeB) "Rodolfo Paoletti", University of Milan, Milan, Italy
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Monica DiLuca
- Department of Pharmacological and Biomolecular Sciences (DiSFeB) "Rodolfo Paoletti", University of Milan, Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences (DiSFeB) "Rodolfo Paoletti", University of Milan, Milan, Italy.
| |
Collapse
|
5
|
A textbook assumption about the brain's most abundant receptors needs to be rewritten. Nature 2025:10.1038/d41586-025-00806-9. [PMID: 40108384 DOI: 10.1038/d41586-025-00806-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
|
6
|
van Oostrum M, Schuman EM. Understanding the molecular diversity of synapses. Nat Rev Neurosci 2025; 26:65-81. [PMID: 39638892 DOI: 10.1038/s41583-024-00888-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Synapses are composed of thousands of proteins, providing the potential for extensive molecular diversity to shape synapse type-specific functional specializations. In this Review, we explore the landscape of synaptic diversity and describe the mechanisms that expand the molecular complexity of synapses, from the genotype to the regulation of gene expression to the production of specific proteoforms and the formation of localized protein complexes. We emphasize the importance of examining every molecular layer and adopting a systems perspective to understand how these interconnected mechanisms shape the diverse functional and structural properties of synapses. We explore current frameworks for classifying synapses and methodologies for investigating different synapse types at varying scales, from synapse-type-specific proteomics to advanced imaging techniques with single-synapse resolution. We highlight the potential of synapse-type-specific approaches for integrating molecular data with cellular functions, circuit organization and organismal phenotypes to enable a more holistic exploration of neuronal phenomena across different scales.
Collapse
Affiliation(s)
- Marc van Oostrum
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
- Biozentrum, University of Basel, Basel, Switzerland
| | - Erin M Schuman
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany.
| |
Collapse
|
7
|
Speca DJ, He CW, Meyer CM, Scott EC, Díaz E. Functional characterization of endocytic signals in the SynDIG/PRRT family members SynDIG1 and SynDIG4 in heterologous cells and neurons. Front Cell Neurosci 2025; 18:1526034. [PMID: 39916936 PMCID: PMC11798926 DOI: 10.3389/fncel.2024.1526034] [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: 11/11/2024] [Accepted: 12/24/2024] [Indexed: 02/09/2025] Open
Abstract
The transmembrane protein Synapse Differentiation Induced Gene 4 (SynDIG4), also known as Proline-rich transmembrane protein 1 (PRRT1), is an AMPA-type glutamate receptor (AMPAR) auxiliary factor that is necessary for maintaining extra-synaptic pools of GluA1. Loss of SynDIG4, and the subsequent decrease in extra-synaptic GluA1, has been found to significantly impact synaptic plasticity in the hippocampus. However, how SynDIG4 establishes and maintains these pools is unclear. Previous studies suggested that endocytic machinery is important for maintaining a pool of mobile surface AMPARs, and that proteins associated with such cellular machinery are critical for proper protein trafficking and internalization. Given that SynDIG4 co-localizes with GluA1 in early and recycling endosomes in cultured hippocampal neurons, we sought to identify the sorting signals that target SynDIG4 to endosomes to further elucidate the role of SynDIG4 in GluA1 trafficking. In this study, we report that SynDIG4 possesses a YxxΦ sorting motif, 178-YVPV-181, responsible for binding to the AP-2 complex cargo-sorting subunit μ2. This motif appears critical for proper SynDIG4 internalization, as SynDIG4 mutant 178-AVPA-181, which disrupts binding to μ2, induces aberrant SynDIG4 accumulation at the plasma-membrane of heterologous cells and primary rat hippocampal neurons. We also show that SynDIG4 mutants lacking an endocytic signal co-localize with GluA1 but less so with GluA2 on the surface of heterologous cells. Furthermore, we show that another family member, SynDIG1, is enriched in the trans-Golgi network (TGN) and can traffic between the TGN and plasma membrane. We have identified a non-canonical μ2 binding sequence in SynDIG1 that induces aberrant accumulation at the plasma membrane of heterologous cells and primary rat hippocampal neurons, suggesting a conserved role for μ2-mediated endocytosis within the SynDIG family. These results provide important insight into the mechanisms by which SynDIG proteins are targeted to endosomal compartments as a step in understanding SynDIG-mediated regulation of AMPAR trafficking.
Collapse
Affiliation(s)
- David J. Speca
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Chun-Wei He
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Christina M. Meyer
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Erin C. Scott
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | - Elva Díaz
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
8
|
de León-López CAM, Carretero-Rey M, Khan ZU. AMPA Receptors in Synaptic Plasticity, Memory Function, and Brain Diseases. Cell Mol Neurobiol 2025; 45:14. [PMID: 39841263 PMCID: PMC11754374 DOI: 10.1007/s10571-024-01529-7] [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: 10/09/2024] [Accepted: 12/26/2024] [Indexed: 01/23/2025]
Abstract
Tetrameric AMPA-type ionotropic glutamate receptors are primary transducers of fast excitatory synaptic transmission in the central nervous system, and their properties and abundance at the synaptic surface are crucial determinants of synaptic efficacy in neuronal communication across the brain. The induction of long-term potentiation (LTP) leads to the insertion of GluA1-containing AMPA receptors at the synaptic surface, whereas during long-term depression (LTD), these receptors are internalized into the cytoplasm of the spine. Disruptions in the trafficking of AMPA receptors to and from the synaptic surface attenuate both forms of synaptic plasticity. Homeostatic scaling up and scaling down, which are additional types of plasticity similar to LTP and LTD, are also regulated by the insertion and removal of GluA1-containing AMPA receptors from the synaptic surface. The trafficking of AMPA receptors is an intricate process assisted by various proteins. Furthermore, AMPA receptors are critical for the formation and consolidation of various types of memory, and alterations in their function are intimately associated with cognitive dysfunction in aging and several neurological and psychiatric diseases. In this review, we will provide an overview of the current understanding of how AMPA receptors regulate various forms of synaptic plasticity, their contribution to memory functions, and their role in aging and brain diseases.
Collapse
Affiliation(s)
- Cristina A Muñoz de León-López
- Laboratory of Neurobiology, Centro de Investigaciones Medico Sanitarias (CIMES), University of Malaga, Calle Marqués de Beccaria, 3, Campus Teatinos s/n, 29010, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Campus Teatinos s/n, Malaga, Spain
| | - Marta Carretero-Rey
- Laboratory of Neurobiology, Centro de Investigaciones Medico Sanitarias (CIMES), University of Malaga, Calle Marqués de Beccaria, 3, Campus Teatinos s/n, 29010, Malaga, Spain
- Department of Medicine, Faculty of Medicine, University of Malaga, Campus Teatinos s/n, Malaga, Spain
| | - Zafar U Khan
- Laboratory of Neurobiology, Centro de Investigaciones Medico Sanitarias (CIMES), University of Malaga, Calle Marqués de Beccaria, 3, Campus Teatinos s/n, 29010, Malaga, Spain.
- Department of Medicine, Faculty of Medicine, University of Malaga, Campus Teatinos s/n, Malaga, Spain.
- CIBERNED, Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
9
|
Lee KS, Lee T, Kim M, Ignatova E, Ban HJ, Sung MK, Kim Y, Kim YJ, Han JH, Choi JK. Shared rare genetic variants in multiplex autism families suggest a social memory gene under selection. Sci Rep 2025; 15:696. [PMID: 39753649 PMCID: PMC11698849 DOI: 10.1038/s41598-024-83839-w] [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: 10/02/2023] [Accepted: 12/17/2024] [Indexed: 01/06/2025] Open
Abstract
Autism spectrum disorder (ASD) affects up to 1 in 59 children, and is one of the most common neurodevelopmental disorders. Recent genomic studies have highlighted the role of rare variants in ASD. This study aimed to identify genes affected by rare variants shared by siblings with ASD and validate the function of a candidate gene FRRS1L. By integrating the whole genome sequencing data of 866 multiplex families from the Hartwell Foundation's Autism Research and Technology Initiative and Autism Speaks MSSNG project, we identified rare variants shared by two or more siblings with ASD. Using shared rare variants (SRVs), we selected candidate genes for ASD. Gene prioritization by evolutionary features and expression alterations on autism identified FRRS1L in two families, including one with impaired social behaviors. One variant in this family was 6 bp away from human-specific trinucleotide fixation. Additionally, CRISPR/Cas9 experiments demonstrated downregulation by a family variant and upregulation by a fixed site. Population genetics further demonstrated that upregulation of this gene has been favored during human evolution. Various mouse behavioral tests showed that Frrs1l knockout specifically impairs social novelty recognition without altering other behavioral phenotypes. Furthermore, we constructed humanized mice by introducing human sequences into a mouse genome. These knockin mice showed improved abilities to retain social memory over time. The results of our population genetic and evolutionary analyses, behavior experiments, and genome editing propose a molecular mechanism that may confer a selective advantage through social memory enhancement and may cause autism-related social impairment when disrupted in humans. These findings highlight the role of FRRS1L, the AMPA receptor subunit, in social behavior and evolution.
Collapse
Affiliation(s)
- Kang Seon Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Taeyeop Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
- Department of Psychiatry, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505, Republic of Korea
- Translational Biomedical Research Group, Asan Institute for Life Science, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Mujun Kim
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea
| | - Elizaveta Ignatova
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Hyo-Jeong Ban
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Min Kyung Sung
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Younghoon Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Youn-Jae Kim
- Specific Organs Cancer Branch, National Cancer Center, Gyeonggi, 10408, Republic of Korea
| | - Jin-Hee Han
- Department of Biological Sciences, KAIST, Daejeon, 34141, Republic of Korea.
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
10
|
Paget-Blanc V, Pronot M, Pfeffer ME, Angelo MF, Herzog E. Purification of Afference-Specific Synaptosome Populations Using Fluorescence-Activated Synaptosome Sorting. Methods Mol Biol 2025; 2910:87-104. [PMID: 40220095 DOI: 10.1007/978-1-0716-4446-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
The central nervous system contains a complex intermingled network of neuronal, glial, and vascular cells, and for several decades, neurobiologists have used subcellular fractionation methods to analyze the molecular structure and functional features of the different cell populations. Biochemists have optimized fractionation protocols that enrich specific compartments such as synapses (called "synaptosomes") and synaptic vesicles to reduce this complexity. However, these approaches suffered from a lack of specificity and purity, which is why we previously extended the conventional synaptosome preparation to purify fluorescent synaptosomes from VGLUT1venus knock-in mice on a cell sorter. We adapted our previous protocol to sort from single neuronal projections and small target regions of the brain as we did in the present example by labeling dopaminergic projections to the striatum. We proved that our newest method allows a steep enrichment in fluorescent dopaminergic synaptosomes containing presynaptic varicosities and associated postsynaptic elements and a substantial depletion in glial contaminants. Here we propose a detailed procedure for implementing projection-specific fluorescence-activated synaptosome sorting.
Collapse
Affiliation(s)
- Vincent Paget-Blanc
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Marie Pronot
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Marlene E Pfeffer
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Maria Florencia Angelo
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Etienne Herzog
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| |
Collapse
|
11
|
Won S, Sweeney CL, Roche KW. Biochemical Properties of Synaptic Proteins Are Dependent on Tissue Preparation: NMDA Receptor Solubility Is Regulated by the C-Terminal Tail. J Cell Biochem 2025; 126:e30664. [PMID: 39370692 PMCID: PMC11730348 DOI: 10.1002/jcb.30664] [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: 07/23/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Synaptic proteins are essential for neuronal development, synaptic transmission, and synaptic plasticity. The postsynaptic density (PSD) is a membrane-associated structure at excitatory synapses, which is composed of a huge protein complex. To understand the interactions and functions of PSD proteins, researchers have employed a variety of imaging and biochemical approaches including sophisticated mass spectrometry. However, the field is lacking a systematic comparison of different experimental conditions and how they might influence the study of the PSD interactome isolated from various tissue preparations. To evaluate the efficiency of several common solubilization conditions, we isolated receptors, scaffolding proteins, and adhesion molecules from brain tissue or primary cultured neurons or human forebrain neurons differentiated from induced pluripotent stem cells (iPSCs). We observed some striking differences in solubility. We found that N-methyl-d-aspartate receptors (NMDARs) and PSD-95 are relatively insoluble in brain tissue, cultured neurons, and human forebrain neurons compared to α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic receptors (AMPARs) or SAP102. In general, synaptic proteins were more soluble in primary neuronal cultures and human forebrain neurons compared to brain tissue. Interestingly, NMDARs are relatively insoluble in HEK293T cells suggesting that insolubility does not directly represent the synaptic fraction but rather it is related to a detergent-insoluble fraction such as lipid rafts. Surprisingly, truncation of the intracellular carboxyl-terminal tail (C-tail) of NMDAR subunits increased NMDAR solubility in HEK293T cells. Our findings show that detergent, pH, and temperature are important for protein preparations to study PSD protein complexes, and NMDAR solubility is regulated by its C-tail, thus providing a technical guide to study synaptic interactomes and subcellular localization of synaptic proteins.
Collapse
Affiliation(s)
- Sehoon Won
- Receptor Biology SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Colin L. Sweeney
- Genetic Immunotherapy SectionNational Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesdaMarylandUSA
| | - Katherine W. Roche
- Receptor Biology SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
12
|
Takato M, Sakamoto S, Nonaka H, Tanimura Valor FY, Tamura T, Hamachi I. Photoproximity labeling of endogenous receptors in the live mouse brain in minutes. Nat Chem Biol 2025; 21:109-119. [PMID: 39090312 DOI: 10.1038/s41589-024-01692-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 07/09/2024] [Indexed: 08/04/2024]
Abstract
Understanding how protein-protein interaction networks in the brain give rise to cognitive functions necessitates their characterization in live animals. However, tools available for this purpose require potentially disruptive genetic modifications and lack the temporal resolution necessary to track rapid changes in vivo. Here we leverage affinity-based targeting and photocatalyzed singlet oxygen generation to identify neurotransmitter receptor-proximal proteins in the live mouse brain using only small-molecule reagents and minutes of photoirradiation. Our photooxidation-driven proximity labeling for proteome identification (named PhoxID) method not only recapitulated the known interactomes of three endogenous neurotransmitter receptors (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR), inhibitory γ-aminobutyric acid type A receptor and ionotropic glutamate receptor delta-2) but also uncovered age-dependent shifts, identifying NECTIN3 and IGSF3 as developmentally regulated AMPAR-proximal proteins in the cerebellum. Overall, this work establishes a flexible and generalizable platform to study receptor microenvironments in genetically intact specimens with an unprecedented temporal resolution.
Collapse
Affiliation(s)
- Mikiko Takato
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Seiji Sakamoto
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
- JST-ERATO, Hamachi Innovative Molecular Technology for Neuroscience, Kyoto, Japan
| | - Hiroshi Nonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
- JST-ERATO, Hamachi Innovative Molecular Technology for Neuroscience, Kyoto, Japan
| | - Fátima Yuri Tanimura Valor
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Tomonori Tamura
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan.
- JST-ERATO, Hamachi Innovative Molecular Technology for Neuroscience, Kyoto, Japan.
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan.
- JST-ERATO, Hamachi Innovative Molecular Technology for Neuroscience, Kyoto, Japan.
| |
Collapse
|
13
|
Leana-Sandoval G, Kolli AV, Chinn CA, Madrid A, Lo I, Sandoval MA, Vera VA, Simms J, Wood MA, Diaz-Alonso J. The GluA1 cytoplasmic tail regulates intracellular AMPA receptor trafficking and synaptic transmission onto dentate gyrus GABAergic interneurons, gating response to novelty. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626277. [PMID: 39677714 PMCID: PMC11643017 DOI: 10.1101/2024.12.01.626277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The GluA1 subunit, encoded by the putative schizophrenia-associated gene GRIA1, is required for activity-regulated AMPA receptor (AMPAR) trafficking, and plays a key role in cognitive and affective function. The cytoplasmic, carboxy-terminal domain (CTD) is the most divergent region across AMPAR subunits. The GluA1 CTD has received considerable attention for its role during long-term potentiation (LTP) at CA1 pyramidal neuron synapses. However, its function at other synapses and, more broadly, its contribution to different GluA1-dependent processes, is poorly understood. Here, we used mice with a constitutive truncation of the GluA1 CTD to dissect its role regulating AMPAR localization and function as well as its contribution to cognitive and affective processes. We found that GluA1 CTD truncation affected AMPAR subunit levels and intracellular trafficking. ΔCTD GluA1 mice exhibited no memory deficits, but presented exacerbated novelty-induced hyperlocomotion and dentate gyrus granule cell (DG GC) hyperactivity, among other behavioral alterations. Mechanistically, we found that AMPAR EPSCs onto DG GABAergic interneurons were significantly reduced, presumably underlying, at least in part, the observed changes in neuronal activity and behavior. In summary, this study dissociates CTD-dependent from CTD-independent GluA1 functions, unveiling the GluA1 CTD as a crucial hub regulating AMPAR function in a cell type-specific manner.
Collapse
Affiliation(s)
- Gerardo Leana-Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Ananth V Kolli
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Carlene A Chinn
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
- Department of Neurobiology & Behavior, University of California at Irvine, CA, 92697, USA
| | - Alexis Madrid
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Iris Lo
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Matthew A Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Vanessa Alizo Vera
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
- Department of Neurobiology & Behavior, University of California at Irvine, CA, 92697, USA
| | - Jeffrey Simms
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Marcelo A Wood
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
- Department of Neurobiology & Behavior, University of California at Irvine, CA, 92697, USA
| | - Javier Diaz-Alonso
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| |
Collapse
|
14
|
Ning L, Shen R, Xie B, Jiang Y, Geng X, Dong W. AMPA receptors in Alzheimer disease: Pathological changes and potential therapeutic targets. J Neuropathol Exp Neurol 2024; 83:895-906. [PMID: 39235983 DOI: 10.1093/jnen/nlae093] [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] [Indexed: 09/07/2024] Open
Abstract
Alzheimer disease (AD) is a prevalent neurodegenerative disorder that affects synapses and leads to progressive cognitive decline. The role of N-methyl-D-aspartic acid (NMDA) receptors in the pathogenesis of AD is well-established as they contribute to excitotoxicity and neurodegeneration in the pathological process of extrasynaptic glutamate concentration. However, the therapeutic potential of the NMDA receptor antagonist memantine in rescuing synaptic damage is limited. Research indicates that α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptors also play a significant role in AD. Abnormal transcription, expression, and localization of AMPA receptors lead to synaptic dysfunction and damage, contributing to early cognitive impairment in AD patients. Understanding the impact of AMPA receptors on AD pathogenesis and exploring the potential for the development of AMPA receptor-targeting drugs are crucial. This review aims to consolidate recent research findings on AMPA receptors in AD, elucidate the current state of AMPA receptor research and lay the foundation for future basic research and drug development.
Collapse
Affiliation(s)
- Luying Ning
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Rongjing Shen
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, Institute of Epigenetics and Brain Science, Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yong Jiang
- Laboratory of Neurological Diseases and Brain Function, Institute of Epigenetics and Brain Science, Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoqi Geng
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
15
|
Sumino A, Sumikama T, Zhao Y, Flechsig H, Umeda K, Kodera N, Konno H, Hattori M, Shibata M. High-Speed Atomic Force Microscopy Reveals Fluctuations and Dimer Splitting of the N-Terminal Domain of GluA2 Ionotropic Glutamate Receptor-Auxiliary Subunit Complex. ACS NANO 2024; 18:25018-25035. [PMID: 39180186 DOI: 10.1021/acsnano.4c06295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazole propionic acid glutamate receptors (AMPARs) enable rapid excitatory synaptic transmission by localizing to the postsynaptic density of glutamatergic spines. AMPARs possess large extracellular N-terminal domains (NTDs), which are crucial for AMPAR clustering at synaptic sites. However, the dynamics of NTDs and the molecular mechanism governing their synaptic clustering remain elusive. Here, we employed high-speed atomic force microscopy (HS-AFM) to directly visualize the conformational dynamics of NTDs in the GluA2 subunit complexed with TARP γ2 in lipid environments. HS-AFM videos of GluA2-γ2 in the resting and activated/open states revealed fluctuations in NTD dimers. Conversely, in the desensitized/closed state, the two NTD dimers adopted a separated conformation with less fluctuation. Notably, we observed individual NTD dimers transitioning into monomers, with extended monomeric states in the activated/open state. Molecular dynamics simulations provided further support, confirming the energetic stability of the monomeric NTD states within lipids. This NTD-dimer splitting resulted in subunit exchange between the receptors and increased the number of interaction sites with synaptic protein neuronal pentraxin 1 (NP1). Moreover, our HS-AFM studies revealed that NP1 forms a ring-shaped octamer through N-terminal disulfide bonds and binds to the tip of the NTD. These findings suggest a molecular mechanism in which NP1, upon forming an octamer, is secreted into the synaptic region and binds to the tip of the GluA2 NTD, thereby bridging and clustering multiple AMPARs. Thus, our findings illuminate the critical role of NTD dynamics in the synaptic clustering of AMPARs and contribute valuable insights into the fundamental processes of synaptic transmission.
Collapse
Affiliation(s)
- Ayumi Sumino
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Takashi Sumikama
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yimeng Zhao
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, and Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Yangpu District, Shanghai 200438, China
- Human Phenome Institute, Fudan University, Yangpu District, Shanghai 200438, China
| | - Holger Flechsig
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Kenichi Umeda
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Noriyuki Kodera
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Hiroki Konno
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Motoyuki Hattori
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Bioactive Small Molecules, Collaborative Innovation Center of Genetics and Development, and Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Yangpu District, Shanghai 200438, China
| | - Mikihiro Shibata
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| |
Collapse
|
16
|
Bats C, Coombs ID, Farrant M, Cull-Candy SG. α-Bungarotoxin labelling of AMPA receptor-associated TARPs in living neurons. Neuroscience 2024:S0306-4522(24)00440-8. [PMID: 39209103 DOI: 10.1016/j.neuroscience.2024.08.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/30/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
In mammalian central neurons AMPARs are clustered at glutamatergic synapses where they mediate fast excitatory transmission. In addition to four pore-forming subunits (GluA1-4), AMPARs contain auxiliary transmembrane AMPAR regulatory proteins (γ2, γ3, γ4, γ5, γ7 or γ8) whose incorporation can vary between neuron types, brain regions, and stages of development. As well as modulating the functional properties of AMPARs, these auxiliary subunits play a central role in AMPAR trafficking. Directly visualizing TARPs could therefore provide a valuable insight into mechanisms underlying these processes. Although antibodies are routinely used for the detection of surface proteins, our experience suggests anti-TARP antibodies are too bulky to access their target, possibly due to close interactions between the extracellular domains of TARP and AMPAR subunits. We therefore assessed the utility of a small monovalent probe - fluorescent α-bungarotoxin (α-Btx) - for TARP labelling in living neurons. We inserted the bungarotoxin binding site (BBS) within the extracellular domain of TARPs to enable their detection in cells exposed to fluorescent α-Btx. Focusing on the prototypical TARP γ2, we demonstrate that the small size of fluorescent α-Btx allows it to bind to the BBS-tagged TARP when associated with AMPARs. Importantly, labelled γ2 enhances AMPAR function in the same way as unmodified γ2. In living neurons, fluorescent α-Btx-labelled γ2 associates with AMPAR clusters at synapses. As a proof-of-principle, we employed our method to compare the surface trafficking of γ2 and γ7 in cerebellar stellate neurons. Our approach provides a simple way to visualize TARPs within AMPARs in living cells.
Collapse
Affiliation(s)
- Cecile Bats
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ian D Coombs
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Mark Farrant
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| | - Stuart G Cull-Candy
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
17
|
Xu QW, Larosa A, Wong TP. Roles of AMPA receptors in social behaviors. Front Synaptic Neurosci 2024; 16:1405510. [PMID: 39056071 PMCID: PMC11269240 DOI: 10.3389/fnsyn.2024.1405510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
As a crucial player in excitatory synaptic transmission, AMPA receptors (AMPARs) contribute to the formation, regulation, and expression of social behaviors. AMPAR modifications have been associated with naturalistic social behaviors, such as aggression, sociability, and social memory, but are also noted in brain diseases featuring impaired social behavior. Understanding the role of AMPARs in social behaviors is timely to reveal therapeutic targets for treating social impairment in disorders, such as autism spectrum disorder and schizophrenia. In this review, we will discuss the contribution of the molecular composition, function, and plasticity of AMPARs to social behaviors. The impact of targeting AMPARs in treating brain disorders will also be discussed.
Collapse
Affiliation(s)
- Qi Wei Xu
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Amanda Larosa
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Tak Pan Wong
- Douglas Hospital Research Centre, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
18
|
Stockwell I, Watson JF, Greger IH. Tuning synaptic strength by regulation of AMPA glutamate receptor localization. Bioessays 2024; 46:e2400006. [PMID: 38693811 PMCID: PMC7616278 DOI: 10.1002/bies.202400006] [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: 01/10/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
Long-term potentiation (LTP) of excitatory synapses is a leading model to explain the concept of information storage in the brain. Multiple mechanisms contribute to LTP, but central amongst them is an increased sensitivity of the postsynaptic membrane to neurotransmitter release. This sensitivity is predominantly determined by the abundance and localization of AMPA-type glutamate receptors (AMPARs). A combination of AMPAR structural data, super-resolution imaging of excitatory synapses, and an abundance of electrophysiological studies are providing an ever-clearer picture of how AMPARs are recruited and organized at synaptic junctions. Here, we review the latest insights into this process, and discuss how both cytoplasmic and extracellular receptor elements cooperate to tune the AMPAR response at the hippocampal CA1 synapse.
Collapse
Affiliation(s)
- Imogen Stockwell
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Jake F. Watson
- Institute of Science and Technology, Technology (IST) Austria, Klosterneuburg, Austria
| | - Ingo H. Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
19
|
Barti B, Dudok B, Kenesei K, Zöldi M, Miczán V, Balla GY, Zala D, Tasso M, Sagheddu C, Kisfali M, Tóth B, Ledri M, Vizi ES, Melis M, Barna L, Lenkei Z, Soltész I, Katona I. Presynaptic nanoscale components of retrograde synaptic signaling. SCIENCE ADVANCES 2024; 10:eado0077. [PMID: 38809980 PMCID: PMC11135421 DOI: 10.1126/sciadv.ado0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024]
Abstract
While our understanding of the nanoscale architecture of anterograde synaptic transmission is rapidly expanding, the qualitative and quantitative molecular principles underlying distinct mechanisms of retrograde synaptic communication remain elusive. We show that a particular form of tonic cannabinoid signaling is essential for setting target cell-dependent synaptic variability. It does not require the activity of the two major endocannabinoid-producing enzymes. Instead, by developing a workflow for physiological, anatomical, and molecular measurements at the same unitary synapse, we demonstrate that the nanoscale stoichiometric ratio of type 1 cannabinoid receptors (CB1Rs) to the release machinery is sufficient to predict synapse-specific release probability. Accordingly, selective decrease of extrasynaptic CB1Rs does not affect synaptic transmission, whereas in vivo exposure to the phytocannabinoid Δ9-tetrahydrocannabinol disrupts the intrasynaptic nanoscale stoichiometry and reduces synaptic variability. These findings imply that synapses leverage the nanoscale stoichiometry of presynaptic receptor coupling to the release machinery to establish synaptic strength in a target cell-dependent manner.
Collapse
Affiliation(s)
- Benjámin Barti
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Barna Dudok
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Departments of Neurology and Neuroscience, Baylor College of Medicine, 1 Baylor Plz, Houston, TX 77030, USA
- Department of Neurosurgery, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA
| | - Kata Kenesei
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Miklós Zöldi
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Vivien Miczán
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Center, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Gyula Y. Balla
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Diana Zala
- Université Paris Cité, INSERM, Institute of Psychiatry and Neurosciences of Paris, F-75014 Paris, France
| | - Mariana Tasso
- Institute of Nanosystems, School of Bio and Nanotechnologies, National University of San Martín - CONICET, 25 de Mayo Ave., 1021 San Martín, Argentina
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, 09042 Cagliari, Italy
| | - Máté Kisfali
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- BiTrial Ltd., Tállya st 23, H-1121 Budapest, Hungary
| | - Blanka Tóth
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Szt. Gellért square 4, H-1111 Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Marco Ledri
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Epilepsy Center, Department of Clinical Sciences, Faculty of Medicine, Lund University, Sölvegatan 17, BMC A11, 221 84 Lund, Sweden
| | - E. Sylvester Vizi
- Molecular Pharmacology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, 09042 Cagliari, Italy
| | - László Barna
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
| | - Zsolt Lenkei
- Université Paris Cité, INSERM, Institute of Psychiatry and Neurosciences of Paris, F-75014 Paris, France
| | - Iván Soltész
- Department of Neurosurgery, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA
| | - István Katona
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| |
Collapse
|
20
|
Targa Dias Anastacio H, Matosin N, Ooi L. Familial Alzheimer's Disease Neurons Bearing Mutations in PSEN1 Display Increased Calcium Responses to AMPA as an Early Calcium Dysregulation Phenotype. Life (Basel) 2024; 14:625. [PMID: 38792645 PMCID: PMC11123496 DOI: 10.3390/life14050625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/18/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Familial Alzheimer's disease (FAD) can be caused by mutations in PSEN1 that encode presenilin-1, a component of the gamma-secretase complex that cleaves amyloid precursor protein. Alterations in calcium (Ca2+) homeostasis and glutamate signaling are implicated in the pathogenesis of FAD; however, it has been difficult to assess in humans whether or not these phenotypes are the result of amyloid or tau pathology. This study aimed to assess the early calcium and glutamate phenotypes of FAD by measuring the Ca2+ response of induced pluripotent stem cell (iPSC)-derived neurons bearing PSEN1 mutations to glutamate and the ionotropic glutamate receptor agonists NMDA, AMPA, and kainate compared to isogenic control and healthy lines. The data show that in early neurons, even in the absence of amyloid and tau phenotypes, FAD neurons exhibit increased Ca2+ responses to glutamate and AMPA, but not NMDA or kainate. Together, this suggests that PSEN1 mutations alter Ca2+ and glutamate signaling as an early phenotype of FAD.
Collapse
Affiliation(s)
- Helena Targa Dias Anastacio
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia;
| | - Natalie Matosin
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia;
| | - Lezanne Ooi
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia;
| |
Collapse
|
21
|
Rinaldi B, Bayat A, Zachariassen LG, Sun JH, Ge YH, Zhao D, Bonde K, Madsen LH, Awad IAA, Bagiran D, Sbeih A, Shah SM, El-Sayed S, Lyngby SM, Pedersen MG, Stenum-Berg C, Walker LC, Krey I, Delahaye-Duriez A, Emrick LT, Sully K, Murali CN, Burrage LC, Plaud Gonzalez JA, Parnes M, Friedman J, Isidor B, Lefranc J, Redon S, Heron D, Mignot C, Keren B, Fradin M, Dubourg C, Mercier S, Besnard T, Cogne B, Deb W, Rivier C, Milani D, Bedeschi MF, Di Napoli C, Grilli F, Marchisio P, Koudijs S, Veenma D, Argilli E, Lynch SA, Au PYB, Ayala Valenzuela FE, Brown C, Masser-Frye D, Jones M, Patron Romero L, Li WL, Thorpe E, Hecher L, Johannsen J, Denecke J, McNiven V, Szuto A, Wakeling E, Cruz V, Sency V, Wang H, Piard J, Kortüm F, Herget T, Bierhals T, Condell A, Ben-Zeev B, Kaur S, Christodoulou J, Piton A, Zweier C, Kraus C, Micalizzi A, Trivisano M, Specchio N, Lesca G, Møller RS, Tümer Z, Musgaard M, Gerard B, Lemke JR, Shi YS, Kristensen AS. Gain-of-function and loss-of-function variants in GRIA3 lead to distinct neurodevelopmental phenotypes. Brain 2024; 147:1837-1855. [PMID: 38038360 PMCID: PMC11068105 DOI: 10.1093/brain/awad403] [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: 07/17/2023] [Revised: 10/17/2023] [Accepted: 11/09/2023] [Indexed: 12/02/2023] Open
Abstract
AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) receptors (AMPARs) mediate fast excitatory neurotransmission in the brain. AMPARs form by homo- or heteromeric assembly of subunits encoded by the GRIA1-GRIA4 genes, of which only GRIA3 is X-chromosomal. Increasing numbers of GRIA3 missense variants are reported in patients with neurodevelopmental disorders (NDD), but only a few have been examined functionally. Here, we evaluated the impact on AMPAR function of one frameshift and 43 rare missense GRIA3 variants identified in patients with NDD by electrophysiological assays. Thirty-one variants alter receptor function and show loss-of-function or gain-of-function properties, whereas 13 appeared neutral. We collected detailed clinical data from 25 patients (from 23 families) harbouring 17 of these variants. All patients had global developmental impairment, mostly moderate (9/25) or severe (12/25). Twelve patients had seizures, including focal motor (6/12), unknown onset motor (4/12), focal impaired awareness (1/12), (atypical) absence (2/12), myoclonic (5/12) and generalized tonic-clonic (1/12) or atonic (1/12) seizures. The epilepsy syndrome was classified as developmental and epileptic encephalopathy in eight patients, developmental encephalopathy without seizures in 13 patients, and intellectual disability with epilepsy in four patients. Limb muscular hypotonia was reported in 13/25, and hypertonia in 10/25. Movement disorders were reported in 14/25, with hyperekplexia or non-epileptic erratic myoclonus being the most prevalent feature (8/25). Correlating receptor functional phenotype with clinical features revealed clinical features for GRIA3-associated NDDs and distinct NDD phenotypes for loss-of-function and gain-of-function variants. Gain-of-function variants were associated with more severe outcomes: patients were younger at the time of seizure onset (median age: 1 month), hypertonic and more often had movement disorders, including hyperekplexia. Patients with loss-of-function variants were older at the time of seizure onset (median age: 16 months), hypotonic and had sleeping disturbances. Loss-of-function and gain-of-function variants were disease-causing in both sexes but affected males often carried de novo or hemizygous loss-of-function variants inherited from healthy mothers, whereas affected females had mostly de novo heterozygous gain-of-function variants.
Collapse
Affiliation(s)
- Berardo Rinaldi
- Medical Genetics Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Allan Bayat
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund 4293, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense 5230Denmark
| | - Linda G Zachariassen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jia-Hui Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Department of Neurology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210032, China
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310030, China
| | - Yu-Han Ge
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Department of Neurology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210032, China
- Ministry of Education Key Laboratory of Model Animal for Disease Study, National Resource Center for Mutant Mice, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210032, China
| | - Dan Zhao
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Kristine Bonde
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Laura H Madsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | | | - Duygu Bagiran
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Amal Sbeih
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Syeda Maidah Shah
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Shaymaa El-Sayed
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Signe M Lyngby
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Miriam G Pedersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Charlotte Stenum-Berg
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Louise Claudia Walker
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Ilona Krey
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig 04103, Germany
| | - Andrée Delahaye-Duriez
- Unité fonctionnelle de médecine génomique et génétique clinique, Hôpital Jean Verdier, Assistance Publique des Hôpitaux de Paris, Bondy 93140, France
- NeuroDiderot, UMR 1141, Inserm, Université Paris Cité, Paris 75019, France
- UFR SMBH, Université Sorbonne Paris Nord, Bobigny 93000, France
| | - Lisa T Emrick
- Division of Neurology and Developmental Neurosciences, Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Krystal Sully
- Division of Neurology and Developmental Neurosciences, Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Chaya N Murali
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Julie Ana Plaud Gonzalez
- Division of Neurology and Developmental Neurosciences, Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Mered Parnes
- Division of Neurology and Developmental Neurosciences, Department of Pediatrics, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
- Pediatric Movement Disorders Clinic, Texas Children’s Hospital and Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennifer Friedman
- Rady Children’s Institute for Genomic Medicine, San Diego, CA 92123, USA
- Department of Neurosciences, University of California San Diego, San Diego, CA 92123, USA
- Department of Pediatrics, University of California San Diego, San Diego, CA 92123, USA
| | - Bertrand Isidor
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes 44000, France
| | - Jérémie Lefranc
- Pediatric Neurophysiology Department, CHU de Brest, Brest 29200, France
| | - Sylvia Redon
- Service de Génétique Médicale, CHU de Brest, Brest 29200, France
- Université de Brest, CHU de Brest, UMR 1078, Brest F29200, France
| | - Delphine Heron
- APHP Sorbonne Université, Département de Génétique, Hôpital Armand Trousseau and Groupe Hospitalier Pitié-Salpêtrière, Paris 75013, France
- Centre de Référence Déficiences Intellectuelles de Causes Rares, Paris 75013, France
| | - Cyril Mignot
- APHP Sorbonne Université, Département de Génétique, Hôpital Armand Trousseau and Groupe Hospitalier Pitié-Salpêtrière, Paris 75013, France
- Centre de Référence Déficiences Intellectuelles de Causes Rares, Paris 75013, France
| | - Boris Keren
- Genetic Department, APHP, Sorbonne Université, Pitié-Salpêtrière Hospital, Paris 75013, France
| | - Mélanie Fradin
- Service de Génétique Médicale, Hôpital Sud, CHU de Rennes, Rennes 35200, France
| | - Christele Dubourg
- Service de Génétique Moléculaire et Génomique, CHU de Rennes, Rennes 35200, France
- Université de Rennes, CNRS, Institut de Genetique et Developpement de Rennes, UMR 6290, Rennes 35200, France
| | - Sandra Mercier
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes 44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes 44000, France
| | - Thomas Besnard
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes 44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes 44000, France
| | - Benjamin Cogne
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes 44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes 44000, France
| | - Wallid Deb
- Nantes Université, CHU Nantes, Service de Génétique Médicale, Nantes 44000, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes 44000, France
| | - Clotilde Rivier
- Department of Paediatrics, Villefranche-sur-Saône Hospital, Villefranche-sur-Saône 69655, France
| | - Donatella Milani
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Maria Francesca Bedeschi
- Medical Genetics Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Claudia Di Napoli
- Medical Genetics Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Federico Grilli
- Medical Genetics Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Paola Marchisio
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pediatria Pneumoinfettivologia, Milan 20122, Italy
- University of Milan, Milan 20122, Italy
| | - Suzanna Koudijs
- Department of Neurology, ENCORE, Erasmus Medical Center-Sophia Children’s Hospital, Rotterdam 3015, The Netherlands
| | - Danielle Veenma
- Department of Pediatrics, ENCORE, Erasmus Medical Center-Sophia Children’s Hospital, Rotterdam 3015, The Netherlands
| | - Emanuela Argilli
- Institute of Human Genetics, University of California, San Francisco, CA 94143, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143, USA
| | - Sally Ann Lynch
- Department of Clinical Genetics, Children’s Health Ireland Crumlin, Dublin D12 N512, Ireland
| | - Ping Yee Billie Au
- Department of Medical Genetics, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | | | - Diane Masser-Frye
- Division of Genetics, Department of Pediatrics, UC San Diego School of Medicine, Rady Children’s Hospital, San Diego, CA 92123, USA
| | - Marilyn Jones
- Division of Genetics, Department of Pediatrics, UC San Diego School of Medicine, Rady Children’s Hospital, San Diego, CA 92123, USA
| | - Leslie Patron Romero
- Facultad de Medicina y Psicología, Universidad Autónoma de Baja California, Tijuana 22010, Mexico
| | | | | | - Laura Hecher
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg 20215, Germany
| | - Jessika Johannsen
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg 20215, Germany
| | - Jonas Denecke
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg 20215, Germany
| | - Vanda McNiven
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1E8, Canada
- Fred A Litwin Family Centre in Genetic Medicine, University Health Network and Mount Sinai Hospital, Toronto, ON M5G 2C4, Canada
| | - Anna Szuto
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1E8, Canada
- Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto, ON M5G 1E8, Canada
| | - Emma Wakeling
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Vincent Cruz
- DDC Clinic Center for Special Needs Children, Middlefield, OH 44062, USA
| | - Valerie Sency
- DDC Clinic Center for Special Needs Children, Middlefield, OH 44062, USA
| | - Heng Wang
- DDC Clinic Center for Special Needs Children, Middlefield, OH 44062, USA
| | - Juliette Piard
- Centre de Génétique Humaine, Centre Hospitalier Universitaire, Université de Franche-Comté, Besançon 25000, France
- UMR 1231 GAD, Inserm, Université de Bourgogne Franche-Comté, Dijon 21000, France
| | - Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Theresia Herget
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Tatjana Bierhals
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Angelo Condell
- Brain and Mitochondrial Research Group, Murdoch Children’s Research Institute, Melbourne, Victoria 3052, Australia
| | - Bruria Ben-Zeev
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan 52621, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv 4R73+8Q, Israel
| | - Simranpreet Kaur
- Brain and Mitochondrial Research Group, Murdoch Children’s Research Institute, Melbourne, Victoria 3052, Australia
- Department of Paediatrics, Melbourne Medical School, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - John Christodoulou
- Brain and Mitochondrial Research Group, Murdoch Children’s Research Institute, Melbourne, Victoria 3052, Australia
- Department of Paediatrics, Melbourne Medical School, University of Melbourne, Melbourne, Victoria 3052, Australia
- Discipline of Genetic Medicine, Sydney Medical School, University of Sydney, Sydney, New South Wales 2050, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NewSouth Wales 2050, Australia
| | - Amelie Piton
- Hôpitaux Universitaires de Strasbourg, Laboratoire de Diagnostic Génétique, Strasbourg 67000, France
| | - Christiane Zweier
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
- Department of Human Genetics, Inselspital Bern, University of Bern, Bern 3010, Switzerland
| | - Cornelia Kraus
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Alessia Micalizzi
- Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Marina Trivisano
- Neurology, Epilepsy and Movement Disorders, Bambino Gesù Children's Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Rome 00165, Italy
| | - Nicola Specchio
- Neurology, Epilepsy and Movement Disorders, Bambino Gesù Children's Hospital, IRCCS, Full Member of European Reference Network EpiCARE, Rome 00165, Italy
| | - Gaetan Lesca
- Department of Medical Genetics, University Hospital of Lyon and Claude Bernard Lyon I University, Lyon 69100, France
- Pathophysiology and Genetics of Neuron and Muscle (PNMG), UCBL, CNRS UMR5261 - INSERM U1315, Lyon 69100, France
| | - Rikke S Møller
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund 4293, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense 5230Denmark
| | - Zeynep Tümer
- Kennedy Center, Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Maria Musgaard
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Benedicte Gerard
- Laboratoires de diagnostic genetique, Institut de genetique Medicale d'Alsace, Hopitaux Universitaires de Strasbourg, Strasbourg 67000, France
| | - Johannes R Lemke
- Center for Rare Diseases, University of Leipzig Medical Center, Leipzig 04103, Germany
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Department of Neurology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210032, China
- Ministry of Education Key Laboratory of Model Animal for Disease Study, National Resource Center for Mutant Mice, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210032, China
- Guangdong Institute of Intelligence Science and Technology, Zhuhai 519031, China
| | - Anders S Kristensen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
22
|
Qneibi M, Bdir S, Bdair M, Aldwaik SA, Sandouka D, Heeh M, Idais TI. AMPA receptor neurotransmission and therapeutic applications: A comprehensive review of their multifaceted modulation. Eur J Med Chem 2024; 266:116151. [PMID: 38237342 DOI: 10.1016/j.ejmech.2024.116151] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 02/05/2024]
Abstract
The neuropharmacological community has shown a strong interest in AMPA receptors as critical components of excitatory synaptic transmission during the last fifteen years. AMPA receptors, members of the ionotropic glutamate receptor family, allow rapid excitatory neurotransmission in the brain. AMPA receptors, which are permeable to sodium and potassium ions, manage the bulk of the brain's rapid synaptic communications. This study thoroughly examines the recent developments in AMPA receptor regulation, focusing on a shift from single chemical illustrations to a more extensive investigation of underlying processes. The complex interplay of these modulators in modifying the function and structure of AMPA receptors is the main focus, providing insight into their influence on the speed of excitatory neurotransmission. This research emphasizes the potential of AMPA receptor modulation as a therapy for various neurological disorders such as epilepsy and Alzheimer's disease. Analyzing these regulators' sophisticated molecular details enhances our comprehension of neuropharmacology, representing a significant advancement in using AMPA receptors for treating intricate neurological conditions.
Collapse
Affiliation(s)
- Mohammad Qneibi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Sosana Bdir
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammad Bdair
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Samia Ammar Aldwaik
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Dana Sandouka
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | | | - Tala Iyad Idais
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| |
Collapse
|
23
|
Zhang T, Dolga AM, Eisel ULM, Schmidt M. Novel crosstalk mechanisms between GluA3 and Epac2 in synaptic plasticity and memory in Alzheimer's disease. Neurobiol Dis 2024; 191:106389. [PMID: 38142840 DOI: 10.1016/j.nbd.2023.106389] [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: 11/23/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease which accounts for the most cases of dementia worldwide. Impaired memory, including acquisition, consolidation, and retrieval, is one of the hallmarks in AD. At the cellular level, dysregulated synaptic plasticity partly due to reduced long-term potentiation (LTP) and enhanced long-term depression (LTD) underlies the memory deficits in AD. GluA3 containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) are one of key receptors involved in rapid neurotransmission and synaptic plasticity. Recent studies revealed a novel form of GluA3 involved in neuronal plasticity that is dependent on cyclic adenosine monophosphate (cAMP), rather than N-methyl-d-aspartate (NMDA). However, this cAMP-dependent GluA3 pathway is specifically and significantly impaired by amyloid beta (Aβ), a pathological marker of AD. cAMP is a key second messenger that plays an important role in modulating memory and synaptic plasticity. We previously reported that exchange protein directly activated by cAMP 2 (Epac2), acting as a main cAMP effector, plays a specific and time-limited role in memory retrieval. From electrophysiological perspective, Epac2 facilities the maintenance of LTP, a cellular event closely associated with memory retrieval. Additionally, Epac2 was found to be involved in the GluA3-mediated plasticity. In this review, we comprehensively summarize current knowledge regarding the specific roles of GluA3 and Epac2 in synaptic plasticity and memory, and their potential association with AD.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9747 AG, Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen 9747 AG, Netherlands
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
24
|
Jimenez-Gomez A, Nguyen MX, Gill JS. Understanding the role of AMPA receptors in autism: insights from circuit and synapse dysfunction. Front Psychiatry 2024; 15:1304300. [PMID: 38352654 PMCID: PMC10861716 DOI: 10.3389/fpsyt.2024.1304300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Autism spectrum disorders represent a diverse etiological spectrum that converge on a syndrome characterized by discrepant deficits in developmental domains often highlighted by concerns in socialization, sensory integration, and autonomic functioning. Importantly, the incidence and prevalence of autism spectrum disorders have seen sharp increases since the syndrome was first described in the 1940s. The wide etiological spectrum and rising number of individuals being diagnosed with the condition lend urgency to capturing a more nuanced understanding of the pathogenic mechanisms underlying the autism spectrum disorders. The current review seeks to understand how the disruption of AMPA receptor (AMPAr)-mediated neurotransmission in the cerebro-cerebellar circuit, particularly in genetic autism related to SHANK3 or SYNGAP1 protein dysfunction function and autism associated with in utero exposure to the anti-seizure medications valproic acid and topiramate, may contribute to the disease presentation. Initially, a discussion contextualizing AMPAr signaling in the cerebro-cerebellar circuitry and microstructural circuit considerations is offered. Subsequently, a detailed review of the literature implicating mutations or deletions of SHANK3 and SYNGAP1 in disrupted AMPAr signaling reveals how bidirectional pathogenic modulation of this key circuit may contribute to autism. Finally, how pharmacological exposure may interact with this pathway, via increased risk of autism diagnosis with valproic acid and topiramate exposure and potential treatment of autism using AMPAr modulator perampanel, is discussed. Through the lens of the review, we will offer speculation on how neuromodulation may be used as a rational adjunct to therapy. Together, the present review seeks to synthesize the disparate considerations of circuit understanding, genetic etiology, and pharmacological modulation to understand the mechanistic interaction of this important and complex disorder.
Collapse
Affiliation(s)
- Andres Jimenez-Gomez
- Neurodevelopmental Disabilities Program, Department of Neurology, Joe DiMaggio Children’s Hospital, Hollywood, FL, United States
| | - Megan X. Nguyen
- Department of Pediatrics, Division of Neurology & Developmental Neurosciences, Baylor College of Medicine, Houston, TX, United States
- Jan & Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Jason S. Gill
- Department of Pediatrics, Division of Neurology & Developmental Neurosciences, Baylor College of Medicine, Houston, TX, United States
- Jan & Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|
25
|
Milham LT, Morris GP, Konen LM, Rentsch P, Avgan N, Vissel B. Quantification of AMPA receptor subunits and RNA editing-related proteins in the J20 mouse model of Alzheimer's disease by capillary western blotting. Front Mol Neurosci 2024; 16:1338065. [PMID: 38299128 PMCID: PMC10828003 DOI: 10.3389/fnmol.2023.1338065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/29/2023] [Indexed: 02/02/2024] Open
Abstract
Introduction Accurate modelling of molecular changes in Alzheimer's disease (AD) dementia is crucial for understanding the mechanisms driving neuronal pathology and for developing treatments. Synaptic dysfunction has long been implicated as a mechanism underpinning memory dysfunction in AD and may result in part from changes in adenosine deaminase acting on RNA (ADAR) mediated RNA editing of the GluA2 subunit of AMPA receptors and changes in AMPA receptor function at the post synaptic cleft. However, few studies have investigated changes in proteins which influence RNA editing and notably, AD studies that focus on studying changes in protein expression, rather than changes in mRNA, often use traditional western blotting. Methods Here, we demonstrate the value of automated capillary western blotting to investigate the protein expression of AMPA receptor subunits (GluA1-4), the ADAR RNA editing proteins (ADAR1-3), and proteins known to regulate RNA editing (PIN1, WWP2, FXR1P, and CREB1), in the J20 AD mouse model. We describe extensive optimisation and validation of the automated capillary western blotting method, demonstrating the use of total protein to normalise protein load, in addition to characterising the optimal protein/antibody concentrations to ensure accurate protein quantification. Following this, we assessed changes in proteins of interest in the hippocampus of 44-week-old J20 AD mice. Results We observed an increase in the expression of ADAR1 p110 and GluA3 and a decrease in ADAR2 in the hippocampus of 44-week-old J20 mice. These changes signify a shift in the balance of proteins that play a critical role at the synapse. Regression analysis revealed unique J20-specific correlations between changes in AMPA receptor subunits, ADAR enzymes, and proteins that regulate ADAR stability in J20 mice, highlighting potential mechanisms mediating RNA-editing changes found in AD. Discussion Our findings in J20 mice generally reflect changes seen in the human AD brain. This study underlines the importance of novel techniques, like automated capillary western blotting, to assess protein expression in AD. It also provides further evidence to support the hypothesis that a dysregulation in RNA editing-related proteins may play a role in the initiation and/or progression of AD.
Collapse
Affiliation(s)
- Luke T. Milham
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Gary P. Morris
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Lyndsey M. Konen
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Peggy Rentsch
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Nesli Avgan
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Bryce Vissel
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
26
|
Yang J, Ma RN, Dong JM, Hu SQ, Liu Y, Yan JZ. Phosphorylation of 4.1N by CaMKII Regulates the Trafficking of GluA1-containing AMPA Receptors During Long-term Potentiation in Acute Rat Hippocampal Brain Slices. Neuroscience 2024; 536:131-142. [PMID: 37993087 DOI: 10.1016/j.neuroscience.2023.11.016] [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: 03/24/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
OBJECTIVE GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (AMPARs) inserted into postsynaptic membranes are key to the process of long-term potentiation (LTP). Some evidence has shown that 4.1N plays a critical role in the membrane trafficking of AMPARs. However, the underlying mechanism behind this is still unclear. We investigated the role of 4.1N-mediated membrane trafficking of AMPARs during theta-burst stimulation long-term potentiation (TBS-LTP), to illustrate the molecular mechanism behind LTP. METHODS LTP was induced by TBS in rat hippocampal CA1 neuron. Tat-GluA1 (MPR), which disrupts the association of 4.1N-GluA1, and autocamtide-2-inhibitory peptide, myristoylated (Myr-AIP), a CaMKII antagonist, were used to explore the role of 4.1N in the AMPARs trafficking during TBS-induced LTP. Immunoprecipitation (IP) and immunoblotting (IB)were used to detect protein expression, phosphorylation, and the interaction of p-CaMKII-4.1N-GluA1. RESULTS We found that Myr-AIP attenuated increases of p-CaMKII (T286), p-GluA1 (ser831), and 4.1N phosphorylation after TBS-LTP, and decreased the association of p-CaMKII-4.1N-GluA1, along with the expression of GluA1, at postsynaptic densities during TBS-LTP. We also designed interfering peptides to disrupt the interaction between 4.1N and GluA1, which showed that Tat-GluA1 (MPR) or Myr-AIP inhibited TBS-LTP and attenuated increases of GluA1 at postsynaptic sites, while Tat-GluA1 (MPR) or Myr-AIP had no effects on miniature excitatory postsynaptic currents (mEPSCs) in non-stimulated hippocampal CA1 neurons. CONCLUSION Active CaMKII enhanced the phosphorylation of 4.1N and facilitated the association of p-CaMKII with 4.1N-GluA1, which in turn resulted in GluA1 trafficking during TBS-LTP. The association of 4.1N-GluA1 is required for LTP, but not for basal synaptic transmission.
Collapse
Affiliation(s)
- Jun Yang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Jiangsu 221004, China
| | - Rui-Ning Ma
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Jiangsu 221004, China
| | - Jia-Min Dong
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Jiangsu 221004, China
| | - Shu-Qun Hu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Jiangsu 221004, China
| | - Yong Liu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Jiangsu 221004, China
| | - Jing-Zhi Yan
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Jiangsu 221004, China.
| |
Collapse
|
27
|
Wu E, Zhang J, Zhang J, Zhu S. Structural insights into gating mechanism and allosteric regulation of NMDA receptors. Curr Opin Neurobiol 2023; 83:102806. [PMID: 37950957 DOI: 10.1016/j.conb.2023.102806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/28/2023] [Accepted: 10/13/2023] [Indexed: 11/13/2023]
Abstract
N-methyl-d-aspartate receptors (NMDARs) belong to the ionotropic glutamate receptors (iGluRs) superfamily and act as coincidence detectors that are crucial to neuronal development and synaptic plasticity. They typically assemble as heterotetramers of two obligatory GluN1 subunits and two alternative GluN2 (from 2A to 2D) and/or GluN3 (3A and 3B) subunits. These alternative subunits mainly determine the diverse biophysical and pharmacological properties of different NMDAR subtypes. Over the past decade, the unprecedented advances in structure elucidation of these tetrameric NMDARs have provided atomic insights into channel gating, allosteric modulation and the action of therapeutic drugs. A wealth of structural and functional information would accelerate the artificial intelligence-based drug design to exploit more NMDAR subtype-specific molecules for the treatment of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Enjiang Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China. https://twitter.com/DuDaDa_Flower
| | - Jilin Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jiwei Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Shujia Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
28
|
Xie RG, Xu GY, Wu SX, Luo C. Presynaptic glutamate receptors in nociception. Pharmacol Ther 2023; 251:108539. [PMID: 37783347 DOI: 10.1016/j.pharmthera.2023.108539] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/19/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Chronic pain is a frequent, distressing and poorly understood health problem. Plasticity of synaptic transmission in the nociceptive pathways after inflammation or injury is assumed to be an important cellular basis for chronic, pathological pain. Glutamate serves as the main excitatory neurotransmitter at key synapses in the somatosensory nociceptive pathways, in which it acts on both ionotropic and metabotropic glutamate receptors. Although conventionally postsynaptic, compelling anatomical and physiological evidence demonstrates the presence of presynaptic glutamate receptors in the nociceptive pathways. Presynaptic glutamate receptors play crucial roles in nociceptive synaptic transmission and plasticity. They modulate presynaptic neurotransmitter release and synaptic plasticity, which in turn regulates pain sensitization. In this review, we summarize the latest understanding of the expression of presynaptic glutamate receptors in the nociceptive pathways, and how they contribute to nociceptive information processing and pain hypersensitivity associated with inflammation / injury. We uncover the cellular and molecular mechanisms of presynaptic glutamate receptors in shaping synaptic transmission and plasticity to mediate pain chronicity, which may provide therapeutic approaches for treatment of chronic pain.
Collapse
Affiliation(s)
- Rou-Gang Xie
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Sheng-Xi Wu
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Ceng Luo
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
29
|
Perozzo AM, Schwenk J, Kamalova A, Nakagawa T, Fakler B, Bowie D. GSG1L-containing AMPA receptor complexes are defined by their spatiotemporal expression, native interactome and allosteric sites. Nat Commun 2023; 14:6799. [PMID: 37884493 PMCID: PMC10603098 DOI: 10.1038/s41467-023-42517-7] [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: 05/15/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
Transmembrane AMPA receptor regulatory proteins (TARPs) and germ cell-specific gene 1-like protein (GSG1L) are claudin-type AMPA receptor (AMPAR) auxiliary subunits that profoundly regulate glutamatergic synapse strength and plasticity. While AMPAR-TARP complexes have been extensively studied, less is known about GSG1L-containing AMPARs. Here, we show that GSG1L's spatiotemporal expression, native interactome and allosteric sites are distinct. GSG1L generally expresses late during brain development in a region-specific manner, constituting about 5% of all AMPAR complexes in adulthood. While GSG1L can co-assemble with TARPs or cornichons (CNIHs), it also assembles as the sole auxiliary subunit. Unexpectedly, GSG1L acts through two discrete evolutionarily-conserved sites on the agonist-binding domain with a weak allosteric interaction at the TARP/KGK site to slow desensitization, and a stronger interaction at a different site that slows recovery from desensitization. Together, these distinctions help explain GSG1L's evolutionary past and how it fulfills a unique signaling role within glutamatergic synapses.
Collapse
Affiliation(s)
- Amanda M Perozzo
- Integrated Program in Neuroscience, McGill University, Montreal, QC, H3A 1A1, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Jochen Schwenk
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104, Freiburg, Germany
| | - Aichurok Kamalova
- Department of Molecular Physiology and Biophysics, Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Terunaga Nakagawa
- Department of Molecular Physiology and Biophysics, Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104, Freiburg, Germany
- Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Derek Bowie
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
30
|
Certain N, Gan Q, Bennett J, Hsieh H, Wollmuth LP. Differential regulation of tetramerization of the AMPA receptor glutamate-gated ion channel by auxiliary subunits. J Biol Chem 2023; 299:105227. [PMID: 37673338 PMCID: PMC10558804 DOI: 10.1016/j.jbc.2023.105227] [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: 02/21/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) auxiliary subunits are specialized, nontransient binding partners of AMPARs that modulate AMPAR channel gating properties and pharmacology, as well as their biogenesis and trafficking. The most well-characterized families of auxiliary subunits are transmembrane AMPAR regulatory proteins (TARPs), cornichon homologs (CNIHs), and the more recently discovered GSG1-L. These auxiliary subunits can promote or reduce surface expression of AMPARs (composed of GluA1-4 subunits) in neurons, thereby impacting their functional role in membrane signaling. Here, we show that CNIH-2 enhances the tetramerization of WT and mutant AMPARs, presumably by increasing the overall stability of the tetrameric complex, an effect that is mainly mediated by interactions with the transmembrane domain of the receptor. We also find CNIH-2 and CNIH-3 show receptor subunit-specific actions in this regard with CNIH-2 enhancing both GluA1 and GluA2 tetramerization, whereas CNIH-3 only weakly enhances GluA1 tetramerization. These results are consistent with the proposed role of CNIHs as endoplasmic reticulum cargo transporters for AMPARs. In contrast, TARP γ-2, TARP γ-8, and GSG1-L have no or negligible effect on AMPAR tetramerization. On the other hand, TARP γ-2 can enhance receptor tetramerization but only when directly fused with the receptor at a maximal stoichiometry. Notably, surface expression of functional AMPARs was enhanced by CNIH-2 to a greater extent than TARP γ-2, suggesting that this distinction aids in maturation and membrane expression. These experiments define a functional distinction between CNIHs and other auxiliary subunits in the regulation of AMPAR biogenesis.
Collapse
Affiliation(s)
- Noele Certain
- Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York, USA
| | - Quan Gan
- Graduate Program in Neuroscience, Stony Brook University, Stony Brook, New York, USA
| | - Joseph Bennett
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, New York, USA
| | - Helen Hsieh
- Department of Surgery, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| | - Lonnie P Wollmuth
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, New York, USA; Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA; Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, New York, USA.
| |
Collapse
|
31
|
Gangwar SP, Yen LY, Yelshanskaya MV, Korman A, Jones DR, Sobolevsky AI. Modulation of GluA2-γ5 synaptic complex desensitization, polyamine block and antiepileptic perampanel inhibition by auxiliary subunit cornichon-2. Nat Struct Mol Biol 2023; 30:1481-1494. [PMID: 37653241 PMCID: PMC10584687 DOI: 10.1038/s41594-023-01080-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 07/26/2023] [Indexed: 09/02/2023]
Abstract
Synaptic complexes of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (AMPARs) with auxiliary subunits mediate most excitatory neurotransmission and can be targeted to treat neuropsychiatric and neurological disorders, including epilepsy. Here we present cryogenic-electron microscopy structures of rat GluA2 AMPAR complexes with inhibitory mouse γ5 and potentiating human cornichon-2 (CNIH2) auxiliary subunits. CNIH2 appears to destabilize the desensitized state of the complex by reducing the separation of the upper lobes in ligand-binding domain dimers. At the same time, CNIH2 stabilizes binding of polyamine spermidine to the selectivity filter of the closed ion channel. Nevertheless, CNIH2, and to a lesser extent γ5, attenuate polyamine block of the open channel and reduce the potency of the antiepileptic drug perampanel that inhibits the synaptic complex allosterically by binding to sites in the ion channel extracellular collar. These findings illustrate the fine-tuning of synaptic complex structure and function in an auxiliary subunit-dependent manner, which is critical for the study of brain region-specific neurotransmission and design of therapeutics for disease treatment.
Collapse
Affiliation(s)
- Shanti Pal Gangwar
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Laura Y Yen
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Cellular and Molecular Physiology and Biophysics Graduate Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Maria V Yelshanskaya
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Aryeh Korman
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
| |
Collapse
|
32
|
Mir A, Amer F, Ali M, Alotaibi W, Alotaibi M, Hedaithy A, Aldurayhim F, Hussain F, Bashir S, Housawi Y. Continuous Spikes and Waves During Sleep (CSWS), Severe Epileptic Encephalopathy, and Choreoathetosis due to Mutations in FRRS1L. Clin EEG Neurosci 2023; 54:526-533. [PMID: 35815844 DOI: 10.1177/15500594221112508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background. Biallelic pathogenic variants in the FRRS1L gene are now known to cause developmental and epileptic encephalopathy-37 (DEE37). It can also be associated with chorea and continuous spikes and waves during sleep (CSWS). CSWS is a rare age-related epileptic encephalopathy syndrome of childhood that is characterized by seizures, neurocognitive regression and electrical status epilepticus during sleep (ESES) on electroencephalogram (EEG) that evolves in four stages. Seizures start during the prodromal phase but the ESES on EEG appears only during acute stage and this is the stage when the diagnosis of CSWS can be made. Methods. We present two patients with FRRS1L mutation causing DEE37 with CSWS. We also review twenty-nine cases of DEE37 described in the literature before and discuss its association with CSWS in the total cohort of thirty-one cases. Results. Developmental regression was found in 80% of the patients, mean age of seizure onset was 18 months, ESES or slow spike and wave on the EEG were reported mostly in the older patients (median age of 11 years) and hypsarrhythmia was reported in younger patients (median age of 4 years). This could suggest that if the younger patients were followed longer their EEG would have evolved into ESES during the acute stage of this syndrome and a diagnosis of CSWS could be made. Conclusion. Recognizing ESES and the natural evolution of CSWS is important in diagnosis and proper management of these patients. More detailed report of EEG findings and the evolution of epilepsy and development are needed to further characterize this syndrome.
Collapse
Affiliation(s)
- Ali Mir
- Department of Pediatric Neurology, King Fahad Specialist Hospital, Dammam, KSA
| | - Fawzia Amer
- Department of Pediatric Neurology, King Fahad Specialist Hospital, Dammam, KSA
- Department of Pediatric Neurology and metabolic, Cairo University Children Hospital, Cairo, Egypt
| | - Mona Ali
- Department of Pediatric Neurology, King Fahad Specialist Hospital, Dammam, KSA
| | - Wajd Alotaibi
- Department of Pediatric Neurology, King Fahad Specialist Hospital, Dammam, KSA
| | - Manar Alotaibi
- Department of Pediatric Neurology, King Fahad Specialist Hospital, Dammam, KSA
| | - Abdullah Hedaithy
- Department of Pediatric Neurology, King Fahad Specialist Hospital, Dammam, KSA
| | - Fatimah Aldurayhim
- Department of Pediatric Neurology, King Fahad Specialist Hospital, Dammam, KSA
| | - Fatimah Hussain
- Department of Pediatric Neurology, King Fahad Specialist Hospital, Dammam, KSA
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital, Dammam, KSA
| | - Yousef Housawi
- Genetic and Metabolic Department, King Fahad Specialist Hospital, Dammam, KSA
| |
Collapse
|
33
|
Boudkkazi S, Schwenk J, Nakaya N, Brechet A, Kollewe A, Harada H, Bildl W, Kulik A, Dong L, Sultana A, Zolles G, Schulte U, Tomarev S, Fakler B. A Noelin-organized extracellular network of proteins required for constitutive and context-dependent anchoring of AMPA-receptors. Neuron 2023; 111:2544-2556.e9. [PMID: 37591201 PMCID: PMC10441612 DOI: 10.1016/j.neuron.2023.07.013] [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: 12/07/2022] [Revised: 04/21/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023]
Abstract
Information processing and storage in the brain rely on AMPA-receptors (AMPARs) and their context-dependent dynamics in synapses and extra-synaptic sites. We found that distribution and dynamics of AMPARs in the plasma membrane are controlled by Noelins, a three-member family of conserved secreted proteins expressed throughout the brain in a cell-type-specific manner. Noelin tetramers tightly assemble with the extracellular domains of AMPARs and interconnect them in a network-like configuration with a variety of secreted and membrane-anchored proteins including Neurexin1, Neuritin1, and Seizure 6-like. Knock out of Noelins1-3 profoundly reduced AMPARs in synapses onto excitatory and inhibitory (inter)neurons, decreased their density and clustering in dendrites, and abolished activity-dependent synaptic plasticity. Our results uncover an endogenous mechanism for extracellular anchoring of AMPARs and establish Noelin-organized networks as versatile determinants of constitutive and context-dependent neurotransmission.
Collapse
Affiliation(s)
- Sami Boudkkazi
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Jochen Schwenk
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Naoki Nakaya
- National Eye Institute, Section of Retinal Ganglion Cell Biology, National Institutes of Health, Bethesda, MD, USA
| | - Aline Brechet
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Astrid Kollewe
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Harumi Harada
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Wolfgang Bildl
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Akos Kulik
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Lijin Dong
- National Eye Institute, Genetic Engineering Facility, National Institutes of Health, Bethesda, MD, USA
| | - Afia Sultana
- National Eye Institute, Section of Retinal Ganglion Cell Biology, National Institutes of Health, Bethesda, MD, USA
| | - Gerd Zolles
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany
| | - Uwe Schulte
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany; Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany; Logopharm GmbH, Schlossstr. 14, 79232 March-Buchheim, Germany
| | - Stanislav Tomarev
- National Eye Institute, Section of Retinal Ganglion Cell Biology, National Institutes of Health, Bethesda, MD, USA.
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, 79104 Freiburg, Germany; Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany.
| |
Collapse
|
34
|
Raven F, Riemersma IW, Olthuis MF, Rybakovaite I, Meijer EL, Meerlo P, Van der Zee EA, Havekes R. Cofilin overactivation improves hippocampus-dependent short-term memory. Front Behav Neurosci 2023; 17:1243524. [PMID: 37638111 PMCID: PMC10448394 DOI: 10.3389/fnbeh.2023.1243524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Many living organisms of the animal kingdom have the fundamental ability to form and retrieve memories. Most information is initially stored as short-term memory, which is then converted to a more stable long-term memory through a process called memory consolidation. At the neuronal level, synaptic plasticity is crucial for memory storage. It includes the formation of new spines, as well as the modification of existing spines, thereby tuning and shaping synaptic efficacy. Cofilin critically contributes to memory processes as upon activation, it regulates the shape of dendritic spines by targeting actin filaments. We previously found that prolonged activation of cofilin in hippocampal neurons attenuated the formation of long-term object-location memories. Because the modification of spine shape and structure is also essential for short-term memory formation, we determined whether overactivation of hippocampal cofilin also influences the formation of short-term memories. To this end, mice were either injected with an adeno-associated virus expressing catalytically active cofilin, or an eGFP control, in the hippocampus. We show for the first time that cofilin overactivation improves short-term memory formation in the object-location memory task, without affecting anxiety-like behavior. Surprisingly, we found no effect of cofilin overactivation on AMPA receptor expression levels. Altogether, while cofilin overactivation might negatively impact the formation of long-lasting memories, it may benefit short-term plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Robbert Havekes
- Neurobiology Expertise Group, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
| |
Collapse
|
35
|
Hoffman JL, Faccidomo SP, Taylor SM, DeMiceli KG, May AM, Smith EN, Whindleton CM, Hodge CW. Negative modulation of AMPA receptors bound to transmembrane AMPA receptor regulatory protein γ-8 blunts the positive reinforcing properties of alcohol and sucrose in a brain region-dependent manner in male mice. Psychopharmacology (Berl) 2023; 240:1261-1273. [PMID: 37055596 PMCID: PMC10698495 DOI: 10.1007/s00213-023-06365-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/06/2023] [Indexed: 04/15/2023]
Abstract
RATIONALE The development and progression of alcohol use disorder (AUD) are widely viewed as maladaptive neuroplasticity. The transmembrane alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) regulatory protein γ8 (TARP γ-8) is a molecular mechanism of neuroplasticity that has not been evaluated in AUD or other addictions. OBJECTIVE To address this gap in knowledge, we evaluated the mechanistic role of TARP γ-8 bound AMPAR activity in the basolateral amygdala (BLA) and ventral hippocampus (vHPC) in the positive reinforcing effects of alcohol, which drive repetitive alcohol use throughout the course of AUD, in male C57BL/6 J mice. These brain regions were selected because they exhibit high levels of TARP γ-8 expression and send glutamate projections to the nucleus accumbens (NAc), which is a key nucleus in the brain reward pathway. METHODS AND RESULTS Site-specific pharmacological inhibition of AMPARs bound to TARP γ-8 in the BLA via bilateral infusion of the selective negative modulator JNJ-55511118 (0-2 µg/µl/side) significantly decreased operant alcohol self-administration with no effect on sucrose self-administration in behavior-matched controls. Temporal analysis showed that reductions in alcohol-reinforced response rate occurred > 25 min after the onset of responding, consistent with a blunting of the positive reinforcing effects of alcohol in the absence of nonspecific behavioral effects. In contrast, inhibition of TARP γ-8 bound AMPARs in the vHPC selectively decreased sucrose self-administration with no effect on alcohol. CONCLUSIONS This study reveals a novel brain region-specific role of TARP γ-8 bound AMPARs as a molecular mechanism of the positive reinforcing effects of alcohol and non-drug rewards.
Collapse
Affiliation(s)
- Jessica L Hoffman
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Thurston-Bowles Building, CB#7178, Chapel Hill, NC, 27599, USA
| | - Sara P Faccidomo
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Thurston-Bowles Building, CB#7178, Chapel Hill, NC, 27599, USA
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Seth M Taylor
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Thurston-Bowles Building, CB#7178, Chapel Hill, NC, 27599, USA
| | - Kristina G DeMiceli
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Thurston-Bowles Building, CB#7178, Chapel Hill, NC, 27599, USA
| | - Ashley M May
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Thurston-Bowles Building, CB#7178, Chapel Hill, NC, 27599, USA
| | - Evan N Smith
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Thurston-Bowles Building, CB#7178, Chapel Hill, NC, 27599, USA
| | - Ciarra M Whindleton
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Thurston-Bowles Building, CB#7178, Chapel Hill, NC, 27599, USA
| | - Clyde W Hodge
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Thurston-Bowles Building, CB#7178, Chapel Hill, NC, 27599, USA.
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
36
|
Alldred MJ, Pidikiti H, Heguy A, Roussos P, Ginsberg SD. Basal forebrain cholinergic neurons are vulnerable in a mouse model of Down syndrome and their molecular fingerprint is rescued by maternal choline supplementation. FASEB J 2023; 37:e22944. [PMID: 37191946 PMCID: PMC10292934 DOI: 10.1096/fj.202202111rr] [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: 12/19/2022] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023]
Abstract
Basal forebrain cholinergic neuron (BFCN) degeneration is a hallmark of Down syndrome (DS) and Alzheimer's disease (AD). Current therapeutics in these disorders have been unsuccessful in slowing disease progression, likely due to poorly understood complex pathological interactions and dysregulated pathways. The Ts65Dn trisomic mouse model recapitulates both cognitive and morphological deficits of DS and AD, including BFCN degeneration and has shown lifelong behavioral changes due to maternal choline supplementation (MCS). To test the impact of MCS on trisomic BFCNs, we performed laser capture microdissection to individually isolate choline acetyltransferase-immunopositive neurons in Ts65Dn and disomic littermates, in conjunction with MCS at the onset of BFCN degeneration. We utilized single population RNA sequencing (RNA-seq) to interrogate transcriptomic changes within medial septal nucleus (MSN) BFCNs. Leveraging multiple bioinformatic analysis programs on differentially expressed genes (DEGs) by genotype and diet, we identified key canonical pathways and altered physiological functions within Ts65Dn MSN BFCNs, which were attenuated by MCS in trisomic offspring, including the cholinergic, glutamatergic and GABAergic pathways. We linked differential gene expression bioinformatically to multiple neurological functions, including motor dysfunction/movement disorder, early onset neurological disease, ataxia and cognitive impairment via Ingenuity Pathway Analysis. DEGs within these identified pathways may underlie aberrant behavior in the DS mice, with MCS attenuating the underlying gene expression changes. We propose MCS ameliorates aberrant BFCN gene expression within the septohippocampal circuit of trisomic mice through normalization of principally the cholinergic, glutamatergic, and GABAergic signaling pathways, resulting in attenuation of underlying neurological disease functions.
Collapse
Affiliation(s)
- Melissa J. Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Harshitha Pidikiti
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
| | - Adriana Heguy
- Genome Technology Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Panos Roussos
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
- Departments of Genetics and Genomic Sciences and Psychiatry and the Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
- Departments of Neuroscience & Physiology, New York University Grossman School of Medicine, New York, NY, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
37
|
Chen ZJ, Su CW, Xiong S, Li T, Liang HY, Lin YH, Chang L, Wu HY, Li F, Zhu DY, Luo CX. Enhanced AMPAR-dependent synaptic transmission by S-nitrosylation in the vmPFC contributes to chronic inflammatory pain-induced persistent anxiety in mice. Acta Pharmacol Sin 2023; 44:954-968. [PMID: 36460834 PMCID: PMC10104852 DOI: 10.1038/s41401-022-01024-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/02/2022] [Indexed: 12/04/2022]
Abstract
Chronic pain patients often have anxiety disorders, and some of them suffer from anxiety even after analgesic administration. In this study, we investigated the role of AMPAR-mediated synaptic transmission in the ventromedial prefrontal cortex (vmPFC) in chronic pain-induced persistent anxiety in mice and explored potential drug targets. Chronic inflammatory pain was induced in mice by bilateral injection of complete Freund's adjuvant (CFA) into the planta of the hind paws; anxiety-like behaviours were assessed with behavioural tests; S-nitrosylation and AMPAR-mediated synaptic transmission were examined using biochemical assays and electrophysiological recordings, respectively. We found that CFA induced persistent upregulation of AMPAR membrane expression and function in the vmPFC of anxious mice but not in the vmPFC of non-anxious mice. The anxious mice exhibited higher S-nitrosylation of stargazin (an AMPAR-interacting protein) in the vmPFC. Inhibition of S-nitrosylation by bilaterally infusing an exogenous stargazin (C302S) mutant into the vmPFC rescued the surface expression of GluA1 and AMPAR-mediated synaptic transmission as well as the anxiety-like behaviours in CFA-injected mice, even after ibuprofen treatment. Moreover, administration of ZL006, a small molecular inhibitor disrupting the interaction of nNOS and PSD-95 (20 mg·kg-1·d-1, for 5 days, i.p.), significantly reduced nitric oxide production and S-nitrosylation of AMPAR-interacting proteins in the vmPFC, resulting in anxiolytic-like effects in anxious mice after ibuprofen treatment. We conclude that S-nitrosylation is necessary for AMPAR trafficking and function in the vmPFC under chronic inflammatory pain-induced persistent anxiety conditions, and nNOS-PSD-95 inhibitors could be potential anxiolytics specific for chronic inflammatory pain-induced persistent anxiety after analgesic treatment.
Collapse
Affiliation(s)
- Zhi-Jin Chen
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Chun-Wan Su
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Shuai Xiong
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Ting Li
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Hai-Ying Liang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- The First Affiliated Hospital of Fujian Medical University, Longyan, 364000, China
| | - Yu-Hui Lin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Lei Chang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Hai-Yin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, China
| | - Fei Li
- Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Dong-Ya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, China
| | - Chun-Xia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, China.
| |
Collapse
|
38
|
Bessa-Neto D, Choquet D. Molecular mechanisms of AMPAR reversible stabilization at synapses. Mol Cell Neurosci 2023; 125:103856. [PMID: 37105372 DOI: 10.1016/j.mcn.2023.103856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
In the central nervous system, glutamatergic synapses play a central role in the regulation of excitatory neuronal transmission. With the membrane-associated guanylate kinase (MAGUK) family of proteins as their structuring scaffold, glutamatergic receptors serve as the powerhouse of glutamatergic synapses. Glutamatergic receptors can be categorized as metabotropic and ionotropic receptors. The latter are then categorized into N-methyl-d-aspartate, kainate receptors, and α-amino-3-hydroxy-5-methyl-isoxazole-propionic acid receptors (AMPARs). Over the past two decades, genetic tagging technology and super-resolution microscopy have been of the utmost importance to unravel how the different receptors are organized at glutamatergic synapses. At the plasma membrane, receptors are highly mobile but show reduced mobility when at synaptic sites. This partial immobilization of receptors at synaptic sites is attributed to the stabilization/anchoring of receptors with the postsynaptic MAGUK proteins and auxiliary proteins, and presynaptic proteins. These partial immobilizations and localization of glutamatergic receptors within the synaptic sites are fundamental for proper basal transmission and synaptic plasticity. Perturbations of the stabilization of glutamatergic receptors are often associated with cognitive deficits. In this review, we describe the proposed mechanisms for synaptic localization and stabilization of AMPARs, the major players of fast excitatory transmission in the central nervous system.
Collapse
Affiliation(s)
- Diogo Bessa-Neto
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Daniel Choquet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France; Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, F-33000 Bordeaux, France.
| |
Collapse
|
39
|
Zhang D, Lape R, Shaikh SA, Kohegyi BK, Watson JF, Cais O, Nakagawa T, Greger IH. Modulatory mechanisms of TARP γ8-selective AMPA receptor therapeutics. Nat Commun 2023; 14:1659. [PMID: 36966141 PMCID: PMC10039940 DOI: 10.1038/s41467-023-37259-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 03/09/2023] [Indexed: 03/27/2023] Open
Abstract
AMPA glutamate receptors (AMPARs) mediate excitatory neurotransmission throughout the brain. Their signalling is uniquely diversified by brain region-specific auxiliary subunits, providing an opportunity for the development of selective therapeutics. AMPARs associated with TARP γ8 are enriched in the hippocampus, and are targets of emerging anti-epileptic drugs. To understand their therapeutic activity, we determined cryo-EM structures of the GluA1/2-γ8 receptor associated with three potent, chemically diverse ligands. We find that despite sharing a lipid-exposed and water-accessible binding pocket, drug action is differentially affected by binding-site mutants. Together with patch-clamp recordings and MD simulations we also demonstrate that ligand-triggered reorganisation of the AMPAR-TARP interface contributes to modulation. Unexpectedly, one ligand (JNJ-61432059) acts bifunctionally, negatively affecting GluA1 but exerting positive modulatory action on GluA2-containing AMPARs, in a TARP stoichiometry-dependent manner. These results further illuminate the action of TARPs, demonstrate the sensitive balance between positive and negative modulatory action, and provide a mechanistic platform for development of both positive and negative selective AMPAR modulators.
Collapse
Affiliation(s)
- Danyang Zhang
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Remigijus Lape
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Saher A Shaikh
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Bianka K Kohegyi
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Jake F Watson
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
- IST Austria, Klosterneuburg, Austria
| | - Ondrej Cais
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Terunaga Nakagawa
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, Nashville, USA
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
40
|
Rao P, Gouaux E. Purification and biochemical analysis of native AMPA receptors from three different mammalian species. PLoS One 2023; 18:e0275351. [PMID: 36930594 PMCID: PMC10022779 DOI: 10.1371/journal.pone.0275351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/29/2022] [Indexed: 03/18/2023] Open
Abstract
The majority of fast, excitatory synaptic transmission in the central nervous system (CNS) is mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), which are glutamate-activated ion channels integral to synaptic plasticity, motor coordination, learning, and memory. Native AMPARs are multiprotein assemblies comprised of a tetrameric receptor core that co-assembles with a broad range of peripheral auxiliary proteins which shape subcellular localization and signaling properties of the resulting complexes. Structure determination of AMPARs has traditionally relied on recombinant expression systems; however, these methods are not well suited to elucidate the diverse array of AMPAR assemblies that are differentially expressed in mammalian brains. While recent studies of native receptor complexes have advanced our understanding of endogenous assemblies, receptors thus far have only been isolated from rodent brain tissue. Here, we employed an immunoaffinity purification strategy to isolate native AMPARs from the brains of three different mammals-pigs, sheep, and cows. Compared to rodents, pigs, sheep, and cows are ungulate mammals, animals with closer genomic identity with humans. Here we determined the molecular size, overall yield, and purity of native AMPARs isolated from these three mammals, thereby demonstrating that structural determination and biochemical analysis is possible from a clade of mammals evolutionarily distinct from rodents.
Collapse
Affiliation(s)
- Prashant Rao
- Vollum Institute, Oregon Health & Science University, Portland, OR, United States of America
| | - Eric Gouaux
- Vollum Institute, Oregon Health & Science University, Portland, OR, United States of America
- Howard Hughes Medical Institute, Oregon Health and Science University, Portland, OR, United States of America
| |
Collapse
|
41
|
The role of post-translational modifications in synaptic AMPA receptor activity. Biochem Soc Trans 2023; 51:315-330. [PMID: 36629507 DOI: 10.1042/bst20220827] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023]
Abstract
AMPA-type receptors for the neurotransmitter glutamate are very dynamic entities, and changes in their synaptic abundance underlie different forms of synaptic plasticity, including long-term synaptic potentiation (LTP), long-term depression (LTD) and homeostatic scaling. The different AMPA receptor subunits (GluA1-GluA4) share a common modular structure and membrane topology, and their intracellular C-terminus tail is responsible for the interaction with intracellular proteins important in receptor trafficking. The latter sequence differs between subunits and contains most sites for post-translational modifications of the receptors, including phosphorylation, O-GlcNAcylation, ubiquitination, acetylation, palmitoylation and nitrosylation, which affect differentially the various subunits. Considering that each single subunit may undergo modifications in multiple sites, and that AMPA receptors may be formed by the assembly of different subunits, this creates multiple layers of regulation of the receptors with impact in synaptic function and plasticity. This review discusses the diversity of mechanisms involved in the post-translational modification of AMPA receptor subunits, and their impact on the subcellular distribution and synaptic activity of the receptors.
Collapse
|
42
|
Certain N, Gan Q, Bennett J, Hsieh H, Wollmuth LP. Differential regulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA) receptor tetramerization by auxiliary subunits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527516. [PMID: 36798164 PMCID: PMC9934675 DOI: 10.1101/2023.02.07.527516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
AMPA receptor (AMPAR) auxiliary subunits are specialized, non-transient binding partners of AMPARs that modulate their ion channel gating properties and pharmacology, as well as their biogenesis and trafficking. The most well characterized families of auxiliary subunits are transmembrane AMPAR regulatory proteins (TARPs) and cornichon homologs (CNIHs) and the more recently discovered GSG1-L. These auxiliary subunits can promote or reduce surface expression of AMPARs in neurons, thereby impacting their functional role in membrane signaling. Here, we show that CNIH-2 enhances the tetramerization of wild type and mutant AMPARs, possibly by increasing the overall stability of the tetrameric complex, an effect that is mainly mediated by interactions with the transmembrane domain of the receptor. We also find CNIH-2 and CNIH-3 show receptor subunit-specific actions in this regard with CNIH-2 enhancing both GluA1 and GluA2 tetramerization whereas CNIH-3 only weakly enhances GluA1 tetramerization. These results are consistent with the proposed role of CNIHs as endoplasmic reticulum cargo transporters for AMPARs. In contrast, TARP γ-2, TARP γ-8, and GSG1-L have no or negligible effect on AMPAR tetramerization. On the other hand, TARP γ-2 can enhance receptor tetramerization but only when directly fused with the receptor at a maximal stoichiometry. Notably, surface expression of functional AMPARs was enhanced by CNIH-2 to a greater extent than TARP γ-2 suggesting that this distinction aids in maturation and membrane expression. These experiments define a functional distinction between CNIHs and other auxiliary subunits in the regulation of AMPAR biogenesis.
Collapse
|
43
|
Okano S, Makita Y, Miyamoto A, Taketazu G, Kimura K, Fukuda I, Tanaka H, Yanagi K, Kaname T. GRIA3 p.Met661Thr variant in a female with developmental epileptic encephalopathy. Hum Genome Var 2023; 10:4. [PMID: 36726007 PMCID: PMC9892509 DOI: 10.1038/s41439-023-00232-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 02/03/2023] Open
Abstract
The X-linked human glutamate receptor subunit 3 (GRIA3) gene (MIM *305915, Xq25) encodes ionotropic α amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA)-type glutamate receptor subunit 3, which mediates postsynaptic neurotransmission. Variants in this gene can cause a variety of neurological disorders, primarily reported in male patients. Here, we report a female patient with developmental and epileptic encephalopathy who carries the novel de novo GRIA3 variant NM_007325.5: c.1982T > C: p.Met661Thr.
Collapse
Affiliation(s)
- Satomi Okano
- Department of Pediatrics, Asahikawa Habilitation Center for Children, Hokkaido, Japan
| | - Yoshio Makita
- Department of Genetic Counseling, Asahikawa Medical University Hospital, Hokkaido, Japan.
| | - Akie Miyamoto
- Department of Pediatrics, Asahikawa Habilitation Center for Children, Hokkaido, Japan
| | - Genya Taketazu
- Department of Pediatrics, Asahikawa Kosei Hospital, Hokkaido, Japan
| | - Kayano Kimura
- Department of Pediatrics, Asahikawa Habilitation Center for Children, Hokkaido, Japan
| | - Ikue Fukuda
- Department of Pediatrics, Asahikawa Habilitation Center for Children, Hokkaido, Japan
| | - Hajime Tanaka
- Department of Pediatrics, Asahikawa Habilitation Center for Children, Hokkaido, Japan
| | - Kumiko Yanagi
- Department of Genome Medicine, National Institute for Child Health and Development, Tokyo, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
44
|
Yamasaki T, Ishii H, Hiraishi A, Kumata K, Wakizaka H, Zhang Y, Kurihara Y, Ogawa M, Nengaki N, Chen J, Li Y, Liang S, Zhang MR. Small-animal PET study for noninvasive quantification of transmembrane AMPA receptor regulatory protein γ-8 (TARP γ-8) in the brain. J Cereb Blood Flow Metab 2023; 43:893-904. [PMID: 36655318 DOI: 10.1177/0271678x231152025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Transmembrane AMPA receptor regulatory protein γ-8 (TARP γ-8) mediates various AMPA receptor functions. Recently, [11C]TARP-2105 was developed as a PET ligand for TARP γ-8 imaging. We performed a full kinetic analysis of [11C]TARP-2105 using PET with [11C]TARP-2105 for the first time. The distribution volume (VT), which is a macro parameter consisting of the K1-k4 rate constants in the two-tissue compartment model analysis, exhibited the following rank order: hippocampus (1.4 ± 0.3) > amygdala (1.0 ± 0.2) > frontal cortex (0.9 ± 0.2) > striatum (0.8 ± 0.2) ≫ cerebellum (0.5 ± 0.1) ≈ thalamus (0.5 ± 0.1) > pons (0.4 ± 0.1 mL/cm3). These heterogenous VT values corresponded with the order of biological distribution of TARP γ-8 in the brain. To validate the reference tissue model, the binding potential (BPND) of [11C]TARP-2105 for TARP γ-8 was estimated using general methods (SRTM, MRTM0, Logan reference model, and ratio method). These BPNDs based on reference models indicated excellent correlation (R2 > 0.9) to the indirect BPNDs based on 2TCM with moderate reproducibility (%variability ≈ 10). PET with [11C]TARP-2105 enabled noninvasive BPND estimation and visual mapping of TARP γ-8 in the living rat brain.
Collapse
Affiliation(s)
- Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Hideki Ishii
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Atsuto Hiraishi
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Katsushi Kumata
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Hidekatsu Wakizaka
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yusuke Kurihara
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan.,SHI Accelerator Service Ltd., Tokyo, Japan
| | - Masanao Ogawa
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan.,SHI Accelerator Service Ltd., Tokyo, Japan
| | - Nobuki Nengaki
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan.,SHI Accelerator Service Ltd., Tokyo, Japan
| | - Jiahui Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston, MA, USA.,Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Yinlong Li
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston, MA, USA.,Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Steven Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston, MA, USA.,Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| |
Collapse
|
45
|
Valdés-Undurraga I, Lobos P, Sánchez-Robledo V, Arias-Cavieres A, SanMartín CD, Barrientos G, More J, Muñoz P, Paula-Lima AC, Hidalgo C, Adasme T. Long-term potentiation and spatial memory training stimulate the hippocampal expression of RyR2 calcium release channels. Front Cell Neurosci 2023; 17:1132121. [PMID: 37025696 PMCID: PMC10071512 DOI: 10.3389/fncel.2023.1132121] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/21/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction: Neuronal Ca2+ signals generated through the activation of Ca2+-induced Ca2+ release in response to activity-generated Ca2+ influx play a significant role in hippocampal synaptic plasticity, spatial learning, and memory. We and others have previously reported that diverse stimulation protocols, or different memory-inducing procedures, enhance the expression of endoplasmic reticulum-resident Ca2+ release channels in rat primary hippocampal neuronal cells or hippocampal tissue. Methods and Results: Here, we report that induction of long-term potentiation (LTP) by Theta burst stimulation protocols of the CA3-CA1 hippocampal synapse increased the mRNA and protein levels of type-2 Ryanodine Receptor (RyR2) Ca2+ release channels in rat hippocampal slices. Suppression of RyR channel activity (1 h preincubation with 20 μM ryanodine) abolished both LTP induction and the enhanced expression of these channels; it also promoted an increase in the surface expression of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits GluR1 and GluR2 and caused a moderate but significant reduction of dendritic spine density. In addition, training rats in the Morris water maze induced memory consolidation, which lasted for several days after the end of the training period, accompanied by an increase in the mRNA levels and the protein content of the RyR2 channel isoform. Discussion: We confirm in this work that LTP induction by TBS protocols requires functional RyR channels. We propose that the increments in the protein content of RyR2 Ca2+ release channels, induced by LTP or spatial memory training, play a significant role in hippocampal synaptic plasticity and spatial memory consolidation.
Collapse
Affiliation(s)
- Ismael Valdés-Undurraga
- Biomedical Research Institute (BNI), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- IVIRMA, Santiago, Chile
| | - Pedro Lobos
- Biomedical Research Institute (BNI), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Center for Advanced Clinical Investigation (CICA), Clinical Hospital, Universidad de Chile, Santiago, Chile
| | | | - Alejandra Arias-Cavieres
- Section of Emergency Medicine, Department of Medicine, Institute for Integrative Physiology, Neuroscience Institute, The University of Chicago, Chicago, IL, United States
| | - Carol D. SanMartín
- Center for Advanced Clinical Investigation (CICA), Clinical Hospital, Universidad de Chile, Santiago, Chile
| | - Genaro Barrientos
- Physiology and Biophysics Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jamileth More
- Center for Advanced Clinical Investigation (CICA), Clinical Hospital, Universidad de Chile, Santiago, Chile
- Laboratory of Translational Psychiatry, Department of Neuroscience and Department de Psychiatry North, Universidad de Chile, Santiago, Chile
| | - Pablo Muñoz
- Translational Neurology Center and Biomedical Research Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile
| | - Andrea Cristina Paula-Lima
- Biomedical Research Institute (BNI), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- Biomedical Research Institute (BNI), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Physiology and Biophysics Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Center for Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Tatiana Adasme
- Biomedical Research Institute (BNI), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Section of Emergency Medicine, Department of Medicine, Institute for Integrative Physiology, Neuroscience Institute, The University of Chicago, Chicago, IL, United States
- Laboratory of Translational Psychiatry, Department of Neuroscience and Department de Psychiatry North, Universidad de Chile, Santiago, Chile
- *Correspondence: Tatiana Adasme
| |
Collapse
|
46
|
Bencsik N, Oueslati Morales CO, Hausser A, Schlett K. Endocytosis of AMPA receptors: Role in neurological conditions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:59-97. [PMID: 36813366 DOI: 10.1016/bs.pmbts.2022.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AMPA receptors are glutamate-gated ion channels, present in a wide range of neuron types and in glial cells. Their main role is to mediate fast excitatory synaptic transmission, and therefore, they are critical for normal brain function. In neurons, AMPA receptors undergo constitutive and activity-dependent trafficking between the synaptic, extrasynaptic and intracellular pools. The kinetics of AMPA receptor trafficking is crucial for the precise functioning of both individual neurons and neural networks involved in information processing and learning. Many of the neurological diseases evoked by neurodevelopmental and neurodegenerative malfunctions or traumatic injuries are caused by impaired synaptic function in the central nervous system. For example, attention-deficit/hyperactivity disorder (ADHD), Alzheimer's disease (AD), tumors, seizures, ischemic strokes, and traumatic brain injury are all characterized by impaired glutamate homeostasis and associated neuronal death, typically caused by excitotoxicity. Given the important role of AMPA receptors in neuronal function, it is not surprising that perturbations in AMPA receptor trafficking are associated with these neurological disorders. In this book chapter, we will first introduce the structure, physiology and synthesis of AMPA receptors, followed by an in-depth description of the molecular mechanisms that control AMPA receptor endocytosis and surface levels under basal conditions or synaptic plasticity. Finally, we will discuss how impairments in AMPA receptor trafficking, particularly endocytosis, contribute to the pathophysiology of various neurological disorders and what efforts are being made to therapeutically target this process.
Collapse
Affiliation(s)
- Norbert Bencsik
- Neuronal Cell Biology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Carlos Omar Oueslati Morales
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Angelika Hausser
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany; Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Katalin Schlett
- Neuronal Cell Biology Research Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
47
|
Coombs ID, Ziobro J, Krotov V, Surtees T, Cull‐Candy SG, Farrant M. A gain-of-function GRIA2 variant associated with neurodevelopmental delay and seizures: Functional characterization and targeted treatment. Epilepsia 2022; 63:e156-e163. [PMID: 36161652 PMCID: PMC10092096 DOI: 10.1111/epi.17419] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 01/11/2023]
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptors (AMPARs) are ligand-gated cationic channels formed from combinations of GluA1-4 subunits. Pathogenic variants of GRIA1-4 have been described in patients with developmental delay, intellectual disability, autism spectrum disorder, and seizures, with GRIA2 variants typically causing AMPAR loss of function. Here, we identify a novel, heterozygous de novo pathogenic missense mutation in GRIA2 (c.1928 C>T, p.A643V, NM_001083619.1) in a 1-year-old boy with epilepsy, developmental delay, and failure to thrive. We made patch-clamp recordings to compare the functional and pharmacological properties of variant and wild-type receptors expressed in HEK293 cells, with and without the transmembrane AMPAR regulatory protein γ2. This showed GluA2 A643V-containing AMPARs to exhibit a novel gain of function, with greatly slowed deactivation, markedly reduced desensitization, and increased glutamate sensitivity. Perampanel, an antiseizure AMPAR negative allosteric modulator, was able to fully block GluA2 A643V/γ2 currents, suggesting potential therapeutic efficacy. The subsequent introduction of perampanel to the patient's treatment regimen was associated with a marked reduction in seizure burden, a resolution of failure to thrive, and clear developmental gains. Our study reveals that GRIA2 disorder can be caused by a gain-of-function variant, and both predicts and suggests the therapeutic efficacy of perampanel. Perampanel may prove beneficial for patients with other gain-of-function GRIA variants.
Collapse
Affiliation(s)
- Ian D. Coombs
- Department of Neuroscience, Physiology, and PharmacologyUniversity College LondonLondonUK
| | - Julie Ziobro
- Department of PediatricsUniversity of MichiganAnn ArborMichiganUSA
| | - Volodymyr Krotov
- Department of Neuroscience, Physiology, and PharmacologyUniversity College LondonLondonUK
| | - Taryn‐Leigh Surtees
- Department of NeurologyWashington University in St Louis School of MedicineSt LouisMissouriUSA
| | - Stuart G. Cull‐Candy
- Department of Neuroscience, Physiology, and PharmacologyUniversity College LondonLondonUK
| | - Mark Farrant
- Department of Neuroscience, Physiology, and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
48
|
van der Spek SJF, Pandya NJ, Koopmans F, Paliukhovich I, van der Schors RC, Otten M, Smit AB, Li KW. Expression and Interaction Proteomics of GluA1- and GluA3-Subunit-Containing AMPARs Reveal Distinct Protein Composition. Cells 2022; 11:cells11223648. [PMID: 36429079 PMCID: PMC9688267 DOI: 10.3390/cells11223648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/27/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
The AMPA glutamate receptor (AMPAR) is the major type of synaptic excitatory ionotropic receptor in the brain. AMPARs have four different subunits, GluA1-4 (each encoded by different genes, Gria1, Gria2, Gria3 and Gria4), that can form distinct tetrameric assemblies. The most abundant AMPAR subtypes in the hippocampus are GluA1/2 and GluA2/3 heterotetramers. Each subtype contributes differentially to mechanisms of synaptic plasticity, which may be in part caused by how these receptors are regulated by specific associated proteins. A broad range of AMPAR interacting proteins have been identified, including the well-studied transmembrane AMPA receptor regulatory proteins TARP-γ2 (also known as Stargazin) and TARP-γ8, Cornichon homolog 2 (CNIH-2) and many others. Several interactors were shown to affect biogenesis, AMPAR trafficking, and channel properties, alone or in distinct assemblies, and several revealed preferred binding to specific AMPAR subunits. To date, a systematic specific interactome analysis of the major GluA1/2 and GluA2/3 AMPAR subtypes separately is lacking. To reveal interactors belonging to specific AMPAR subcomplexes, we performed both expression and interaction proteomics on hippocampi of wildtype and Gria1- or Gria3 knock-out mice. Whereas GluA1/2 receptors co-purified TARP-γ8, synapse differentiation-induced protein 4 (SynDIG4, also known as Prrt1) and CNIH-2 with highest abundances, GluA2/3 receptors revealed strongest co-purification of CNIH-2, TARP-γ2, and Noelin1 (or Olfactomedin-1). Further analysis revealed that TARP-γ8-SynDIG4 interact directly and co-assemble into an AMPAR subcomplex especially at synaptic sites. Together, these data provide a framework for further functional analysis into AMPAR subtype specific pathways in health and disease.
Collapse
|
49
|
Zeppillo T, Schulmann A, Macciardi F, Hjelm BE, Föcking M, Sequeira PA, Guella I, Cotter D, Bunney WE, Limon A, Vawter MP. Functional impairment of cortical AMPA receptors in schizophrenia. Schizophr Res 2022; 249:25-37. [PMID: 32513544 PMCID: PMC7718399 DOI: 10.1016/j.schres.2020.03.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/14/2022]
Abstract
Clinical and preclinical studies suggest that some of the behavioral alterations observed in schizophrenia (SZ) may be mechanistically linked to synaptic dysfunction of glutamatergic signaling. Recent genetic and proteomic studies suggest alterations of cortical glutamate receptors of the AMPA-type (AMPARs), which are the predominant ligand-gated ionic channels of fast transmission at excitatory synapses. The impact of gene and protein alterations on the electrophysiological activity of AMPARs is not known in SZ. In this proof of principle work, using human postmortem brain synaptic membranes isolated from the dorsolateral prefrontal cortex (DLPFC), we combined electrophysiological analysis from microtransplanted synaptic membranes (MSM) with transcriptomic (RNA-Seq) and label-free proteomics data in 10 control and 10 subjects diagnosed with SZ. We observed in SZ a reduction in the amplitude of AMPARs currents elicited by kainate, an agonist of AMPARs that blocks the desensitization of the receptor. This reduction was not associated with protein abundance but with a reduction in kainate's potency to activate AMPARs. Electrophysiologically-anchored dataset analysis (EDA) was used to identify synaptosomal proteins that linearly correlate with the amplitude of the AMPARs responses, gene ontology functional annotations were then used to determine protein-protein interactions. Protein modules associated with positive AMPARs current increases were downregulated in SZ, while protein modules that were upregulated in SZ were associated with decreased AMPARs currents. Our results indicate that transcriptomic and proteomic alterations, frequently observed in the DLPFC in SZ, converge at the synaptic level producing a functional electrophysiological impairment of AMPARs.
Collapse
Affiliation(s)
- Tommaso Zeppillo
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch at Galveston, USA; Department of Life Sciences, University of Trieste, B.R.A.I.N., Centre for Neuroscience, Trieste, Italy
| | - Anton Schulmann
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA; Current address: National Institute of Mental Health, Human Genetics Branch, Bethesda, MD, USA
| | - Fabio Macciardi
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA
| | - Brooke E Hjelm
- Department of Translational Genomics, Keck School of Medicine of USC, University of Southern California (USC), Los Angeles, CA, USA
| | | | - P Adolfo Sequeira
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA
| | - Ilaria Guella
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA
| | - David Cotter
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - William E Bunney
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA
| | - Agenor Limon
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, School of Medicine, University of Texas Medical Branch at Galveston, USA.
| | - Marquis P Vawter
- Department of Psychiatry & Human Behavior, University of California Irvine, CA 92697, USA.
| |
Collapse
|
50
|
Wu QL, Gao Y, Li JT, Ma WY, Chen NH. The Role of AMPARs Composition and Trafficking in Synaptic Plasticity and Diseases. Cell Mol Neurobiol 2022; 42:2489-2504. [PMID: 34436728 PMCID: PMC11421597 DOI: 10.1007/s10571-021-01141-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/11/2021] [Indexed: 11/28/2022]
Abstract
AMPA receptors are tetrameric ionic glutamate receptors, which mediate 90% fast excitatory synaptic transmission induced by excitatory glutamate in the mammalian central nervous system through the activation or inactivation of ion channels. The alternation of synaptic AMPA receptor number and subtype is thought to be one of the primary mechanisms that involve in synaptic plasticity regulation and affect the functions in learning, memory, and cognition. The increasing of surface AMPARs enhances synaptic strength during long-term potentiation, whereas the decreasing of AMPARs weakens synaptic strength during the long-term depression. It is closely related to the AMPA receptor as well as its subunits assembly, trafficking, and degradation. The dysfunction of any step in these precise regulatory processes is likely to induce the disorder of synaptic transmission and loss of neurons, or even cause neuropsychiatric diseases ultimately. Therefore, it is useful to understand how AMPARs regulate synaptic plasticity and its role in related neuropsychiatric diseases via comprehending architecture and trafficking of the receptors. Here, we reviewed the progress in structure, expression, trafficking, and relationship with synaptic plasticity of AMPA receptor, especially in anxiety, depression, neurodegenerative disorders, and cerebral ischemia.
Collapse
Affiliation(s)
- Qing-Lin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yan Gao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jun-Tong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Yu Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nai-Hong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|