1
|
Pan G, Chai L, Chen R, Yuan Q, Song Z, Feng W, Wei J, Yang Z, Zhang Y, Xie G, Yan A, Lv Q, Wang C, Zhao Y, Wang Y. Potential mechanism of Qinggong Shoutao pill alleviating age-associated memory decline based on integration strategy. PHARMACEUTICAL BIOLOGY 2024; 62:105-119. [PMID: 38145345 PMCID: PMC10763866 DOI: 10.1080/13880209.2023.2291689] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 11/30/2023] [Indexed: 12/26/2023]
Abstract
CONTEXT Qinggong Shoutao Wan (QGSTW) is a pill used as a traditional medicine to treat age-associated memory decline (AAMI). However, its potential mechanisms are unclear. OBJECTIVE This study elucidates the possible mechanisms of QGSTW in treating AAMI. MATERIALS AND METHODS Network pharmacology and molecular docking approaches were utilized to identify the potential pathway by which QGSTW alleviates AAMI. C57BL/6J mice were divided randomly into control, model, and QGSTW groups. A mouse model of AAMI was established by d-galactose, and the pathways that QGSTW acts on to ameliorate AAMI were determined by ELISA, immunofluorescence staining and Western blotting after treatment with d-gal (100 mg/kg) and QGSTW (20 mL/kg) for 12 weeks. RESULTS Network pharmacology demonstrated that the targets of the active components were significantly enriched in the cAMP signaling pathway. AKT1, FOS, GRIN2B, and GRIN1 were the core target proteins. QGSTW treatment increased the discrimination index from -16.92 ± 7.06 to 23.88 ± 15.94% in the novel location test and from -19.54 ± 5.71 to 17.55 ± 6.73% in the novel object recognition test. ELISA showed that QGSTW could increase the levels of cAMP. Western blot analysis revealed that QGSTW could upregulate the expression of PKA, CREB, c-Fos, GluN1, GluA1, CaMKII-α, and SYN. Immunostaining revealed that the expression of SYN was decreased in the CA1 and DG. DISCUSSION AND CONCLUSIONS This study not only provides new insights into the mechanism of QGSTW in the treatment of AAMI but also provides important information and new research ideas for the discovery of traditional Chinese medicine compounds that can treat AAMI.
Collapse
Affiliation(s)
- Guiyun Pan
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lijuan Chai
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Yuan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Song
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wanying Feng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinna Wei
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihua Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhang Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guinan Xie
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - An Yan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingbo Lv
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Caijun Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingqiang Zhao
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
2
|
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
|
3
|
Cassel JC, Panzer E, Guimaraes-Olmo I, Cosquer B, de Vasconcelos AP, Stephan A. The ventral midline thalamus and long-term memory: What consolidation, what retrieval, what plasticity in rodents? Neurosci Biobehav Rev 2024; 167:105932. [PMID: 39454977 DOI: 10.1016/j.neubiorev.2024.105932] [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: 07/18/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
The ventral midline thalamus, including the reuniens and rhomboid (ReRh) nuclei, connects bidirectionally with the medial prefrontal cortex (mPFC) and hippocampus (Hip), both essential for memory processes. This review compiles and discusses studies on a role for the ReRh nuclei in the system consolidation of memories, also considering their potentially limited participation in memory retrieval or early phases of consolidation. It also examines scientific literature on short- and long-term plasticity in ReRh-mPFC and ReRh-Hip connections, emphasizing plasticity's importance in understanding these nuclei's role in memory. The idea that the two nuclei are at the crossroads of information exchange between the mPFC and the Hip is not new, but the relationship between this status and the plasticity of their connections remains elusive. Since this perspective is relatively recent, our concluding section suggests conceptual and practical avenues for future research, aiming perhaps to bring more order to the apparently multi-functional implication of the ventral midline thalamus in cognition.
Collapse
Affiliation(s)
- Jean-Christophe Cassel
- Laboratoire de Neurosciences Cognitives et Adaptatives, Université de Strasbourg, Strasbourg 67000, France; LNCA, UMR 7364 - CNRS, Strasbourg 67000, France.
| | - Elodie Panzer
- Laboratoire de Neurosciences Cognitives et Adaptatives, Université de Strasbourg, Strasbourg 67000, France; LNCA, UMR 7364 - CNRS, Strasbourg 67000, France
| | - Isabella Guimaraes-Olmo
- Laboratoire de Neurosciences Cognitives et Adaptatives, Université de Strasbourg, Strasbourg 67000, France; LNCA, UMR 7364 - CNRS, Strasbourg 67000, France
| | - Brigitte Cosquer
- Laboratoire de Neurosciences Cognitives et Adaptatives, Université de Strasbourg, Strasbourg 67000, France; LNCA, UMR 7364 - CNRS, Strasbourg 67000, France
| | - Anne Pereira de Vasconcelos
- Laboratoire de Neurosciences Cognitives et Adaptatives, Université de Strasbourg, Strasbourg 67000, France; LNCA, UMR 7364 - CNRS, Strasbourg 67000, France
| | - Aline Stephan
- Laboratoire de Neurosciences Cognitives et Adaptatives, Université de Strasbourg, Strasbourg 67000, France; LNCA, UMR 7364 - CNRS, Strasbourg 67000, France
| |
Collapse
|
4
|
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 2024. [PMID: 39394632 DOI: 10.1111/febs.17287] [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: 05/18/2024] [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
|
5
|
How protons shape AMPA receptor structure, function and diffusion at the synapse. Nat Struct Mol Biol 2024; 31:1466-1467. [PMID: 39138331 DOI: 10.1038/s41594-024-01371-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
|
6
|
Tropea MR, Melone M, Li Puma DD, Vacanti V, Aceto G, Bandiera B, Trovato RC, Torrisi SA, Leggio GM, Palmeri A, D'Ascenzo M, Conti F, Grassi C, Puzzo D. Blockade of dopamine D3 receptors improves hippocampal synaptic function and rescues age-related cognitive phenotype. Aging Cell 2024:e14291. [PMID: 39236310 DOI: 10.1111/acel.14291] [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: 03/01/2024] [Revised: 06/22/2024] [Accepted: 07/16/2024] [Indexed: 09/07/2024] Open
Abstract
Dopamine D3 receptors (D3Rs) modulate neuronal activity in several brain regions including the hippocampus. Although previous studies reported that blocking D3Rs exerts pro-cognitive effects, their involvement in hippocampal synaptic function and memory in the healthy and aged brain has not been thoroughly investigated. We demonstrated that in adult wild type (WT) mice, D3R pharmacological blockade or genetic deletion as in D3 knock out (KO) mice, converted the weak form of long-term potentiation (LTP1) into the stronger long-lasting LTP (LTP2) via the cAMP/PKA pathway, and allowed the formation of long-term memory. D3R effects were mainly mediated by post-synaptic mechanisms as their blockade enhanced basal synaptic transmission (BST), AMPAR-mediated currents, mEPSC amplitude, and the expression of the post-synaptic proteins PSD-95, phospho(p)GluA1 and p-CREB. Consistently, electron microscopy revealed a prevalent expression of D3Rs in post-synaptic dendrites. Interestingly, with age, D3Rs decreased in axon terminals while maintaining their levels in post-synaptic dendrites. Indeed, in aged WT mice, blocking D3Rs reversed the impairment of LTP, BST, memory, post-synaptic protein expression, and PSD length. Notably, aged D3-KO mice did not exhibit synaptic and memory deficits. In conclusion, we demonstrated the fundamental role of D3Rs in hippocampal synaptic function and memory, and their potential as a therapeutic target to counteract the age-related hippocampal cognitive decline.
Collapse
Affiliation(s)
- Maria Rosaria Tropea
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Marcello Melone
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, Ancona, Italy
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Valeria Vacanti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Aceto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Bruno Bandiera
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberta Carmela Trovato
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Gian Marco Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Agostino Palmeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Marcello D'Ascenzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica Delle Marche, Ancona, Italy
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Oasi Research Institute-IRCCS, Troina, Italy
| |
Collapse
|
7
|
Harris KM, Kuwajima M, Flores JC, Zito K. Synapse-specific structural plasticity that protects and refines local circuits during LTP and LTD. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230224. [PMID: 38853547 PMCID: PMC11529630 DOI: 10.1098/rstb.2023.0224] [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: 10/17/2023] [Revised: 12/20/2023] [Accepted: 01/05/2024] [Indexed: 06/11/2024] Open
Abstract
Synapses form trillions of connections in the brain. Long-term potentiation (LTP) and long-term depression (LTD) are cellular mechanisms vital for learning that modify the strength and structure of synapses. Three-dimensional reconstruction from serial section electron microscopy reveals three distinct pre- to post-synaptic arrangements: strong active zones (AZs) with tightly docked vesicles, weak AZs with loose or non-docked vesicles, and nascent zones (NZs) with a postsynaptic density but no presynaptic vesicles. Importantly, LTP can be temporarily saturated preventing further increases in synaptic strength. At the onset of LTP, vesicles are recruited to NZs, converting them to AZs. During recovery of LTP from saturation (1-4 h), new NZs form, especially on spines where AZs are most enlarged by LTP. Sentinel spines contain smooth endoplasmic reticulum (SER), have the largest synapses and form clusters with smaller spines lacking SER after LTP recovers. We propose a model whereby NZ plasticity provides synapse-specific AZ expansion during LTP and loss of weak AZs that drive synapse shrinkage during LTD. Spine clusters become functionally engaged during LTP or disassembled during LTD. Saturation of LTP or LTD probably acts to protect recently formed memories from ongoing plasticity and may account for the advantage of spaced over massed learning. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Kristen M. Harris
- Department of Neuroscience and Center for Learning and Memory, The University of Texas at Austin, Austin, TX78712, USA
| | - Masaaki Kuwajima
- Department of Neuroscience and Center for Learning and Memory, The University of Texas at Austin, Austin, TX78712, USA
| | - Juan C. Flores
- Center for Neuroscience, University of California, Davis, CA95618, USA
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, CA95618, USA
| |
Collapse
|
8
|
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
|
9
|
Singh N, Nandy SK, Jyoti A, Saxena J, Sharma A, Siddiqui AJ, Sharma L. Protein Kinase C (PKC) in Neurological Health: Implications for Alzheimer's Disease and Chronic Alcohol Consumption. Brain Sci 2024; 14:554. [PMID: 38928554 PMCID: PMC11201589 DOI: 10.3390/brainsci14060554] [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: 05/07/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Protein kinase C (PKC) is a diverse enzyme family crucial for cell signalling in various organs. Its dysregulation is linked to numerous diseases, including cancer, cardiovascular disorders, and neurological problems. In the brain, PKC plays pivotal roles in synaptic plasticity, learning, memory, and neuronal survival. Specifically, PKC's involvement in Alzheimer's Disease (AD) pathogenesis is of significant interest. The dysregulation of PKC signalling has been linked to neurological disorders, including AD. This review elucidates PKC's pivotal role in neurological health, particularly its implications in AD pathogenesis and chronic alcohol addiction. AD, characterised by neurodegeneration, implicates PKC dysregulation in synaptic dysfunction and cognitive decline. Conversely, chronic alcohol consumption elicits neural adaptations intertwined with PKC signalling, exacerbating addictive behaviours. By unravelling PKC's involvement in these afflictions, potential therapeutic avenues emerge, offering promise for ameliorating their debilitating effects. This review navigates the complex interplay between PKC, AD pathology, and alcohol addiction, illuminating pathways for future neurotherapeutic interventions.
Collapse
Affiliation(s)
- Nishtha Singh
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| | - Shouvik Kumar Nandy
- School of Pharmacy, Techno India University, Sector-V, Kolkata 700091, West Bengal, India;
| | - Anupam Jyoti
- Department of Life Science, Parul Institute of Applied Science, Parul University, Vadodara 391760, Gujarat, India;
| | - Juhi Saxena
- Department of Biotechnology, Parul Institute of Technology, Parul University, Vadodara 391760, Gujarat, India;
| | - Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail 55476, Saudi Arabia
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology, and Management Sciences, Solan 173229, Himachal Pradesh, India; (N.S.); (A.S.)
| |
Collapse
|
10
|
Cai L, Xu J, Liu J, Luo H, Yang R, Gui X, Wei L. miRNAs in treatment-resistant depression: a systematic review. Mol Biol Rep 2024; 51:638. [PMID: 38727891 DOI: 10.1007/s11033-024-09554-x] [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: 12/24/2023] [Accepted: 04/15/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Treatment-resistant depression (TRD) is a condition in a subset of depressed patients characterized by resistance to antidepressant medications. The global prevalence of TRD has been steadily increasing, yet significant advancements in its diagnosis and treatment remain elusive despite extensive research efforts. The precise underlying pathogenic mechanisms are still not fully understood. Epigenetic mechanisms play a vital role in a wide range of diseases. In recent years, investigators have increasingly focused on the regulatory roles of miRNAs in the onset and progression of TRD. miRNAs are a class of noncoding RNA molecules that regulate the translation and degradation of their target mRNAs via interaction, making the exploration of their functions in TRD essential for elucidating their pathogenic mechanisms. METHODS AND RESULTS A systematic search was conducted in four databases, namely PubMed, Web of Science, Cochrane Library, and Embase, focusing on studies related to treatment-resistant depression and miRNAs. The search was performed using terms individually or in combination, such as "treatment-resistant depression," "medication-resistant depression," and "miRNAs." The selected articles were reviewed and collated, covering the time period from the inception of each database to the end of February 2024. We found that miRNAs play a crucial role in the pathophysiology of TRD through three main aspects: 1) involvement in miRNA-mediated inflammatory responses (including miR-155, miR-345-5p, miR-146a, and miR-146a-5p); 2) influence on 5-HT transport processes (including miR-674,miR-708, and miR-133a); and 3) regulation of synaptic plasticity (including has-miR-335-5p,has-miR- 1292-3p, let-7b, and let-7c). Investigating the differential expression and interactions of these miRNAs could contribute to a deeper understanding of the molecular mechanisms underlying TRD. CONCLUSIONS miRNAs might play a pivotal role in the pathogenesis of TRD. Gaining a deeper understanding of the roles and interrelations of miRNAs in TRD will contribute to elucidating disease pathogenesis and potentially provide avenues for the development of novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Lun Cai
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530023, People's Republic of China
| | - Jingwen Xu
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530023, People's Republic of China
| | - Jie Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530023, People's Republic of China
| | - Huazheng Luo
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530023, People's Republic of China
| | - Rongrong Yang
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530023, People's Republic of China
| | - Xiongbin Gui
- Department of Surgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Nanning, 530000, Guangxi, People's Republic of China.
| | - Liping Wei
- Department of Surgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, No. 89-9 Dongge Road, Nanning, 530000, Guangxi, People's Republic of China
| |
Collapse
|
11
|
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
|
12
|
McFarlan AR, Guo C, Gomez I, Weinerman C, Liang TA, Sjöström PJ. The spike-timing-dependent plasticity of VIP interneurons in motor cortex. Front Cell Neurosci 2024; 18:1389094. [PMID: 38706517 PMCID: PMC11066220 DOI: 10.3389/fncel.2024.1389094] [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: 02/20/2024] [Accepted: 04/09/2024] [Indexed: 05/07/2024] Open
Abstract
The plasticity of inhibitory interneurons (INs) plays an important role in the organization and maintenance of cortical microcircuits. Given the many different IN types, there is an even greater diversity in synapse-type-specific plasticity learning rules at excitatory to excitatory (E→I), I→E, and I→I synapses. I→I synapses play a key disinhibitory role in cortical circuits. Because they typically target other INs, vasoactive intestinal peptide (VIP) INs are often featured in I→I→E disinhibition, which upregulates activity in nearby excitatory neurons. VIP IN dysregulation may thus lead to neuropathologies such as epilepsy. In spite of the important activity regulatory role of VIP INs, their long-term plasticity has not been described. Therefore, we characterized the phenomenology of spike-timing-dependent plasticity (STDP) at inputs and outputs of genetically defined VIP INs. Using a combination of whole-cell recording, 2-photon microscopy, and optogenetics, we explored I→I STDP at layer 2/3 (L2/3) VIP IN outputs onto L5 Martinotti cells (MCs) and basket cells (BCs). We found that VIP IN→MC synapses underwent causal long-term depression (LTD) that was presynaptically expressed. VIP IN→BC connections, however, did not undergo any detectable plasticity. Conversely, using extracellular stimulation, we explored E→I STDP at inputs to VIP INs which revealed long-term potentiation (LTP) for both causal and acausal timings. Taken together, our results demonstrate that VIP INs possess synapse-type-specific learning rules at their inputs and outputs. This suggests the possibility of harnessing VIP IN long-term plasticity to control activity-related neuropathologies such as epilepsy.
Collapse
Affiliation(s)
- Amanda R. McFarlan
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Connie Guo
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Isabella Gomez
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Chaim Weinerman
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Tasha A. Liang
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - P. Jesper Sjöström
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| |
Collapse
|
13
|
Midorikawa R, Wakazono Y, Takamiya K. Aβ peptide enhances GluA1 internalization via lipid rafts in Alzheimer's-related hippocampal LTP dysfunction. J Cell Sci 2024; 137:jcs261281. [PMID: 38668720 DOI: 10.1242/jcs.261281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 03/08/2024] [Indexed: 05/01/2024] Open
Abstract
Amyloid β (Aβ) is a central contributor to neuronal damage and cognitive impairment in Alzheimer's disease (AD). Aβ disrupts AMPA receptor-mediated synaptic plasticity, a key factor in early AD progression. Numerous studies propose that Aβ oligomers hinder synaptic plasticity, particularly long-term potentiation (LTP), by disrupting GluA1 (encoded by GRIA1) function, although the precise mechanism remains unclear. In this study, we demonstrate that Aβ mediates the accumulation of GM1 ganglioside in lipid raft domains of cultured cells, and GluA1 exhibits preferential localization in lipid rafts via direct binding to GM1. Aβ enhances the raft localization of GluA1 by increasing GM1 in these areas. Additionally, chemical LTP stimulation induces lipid raft-dependent GluA1 internalization in Aβ-treated neurons, resulting in reduced cell surface and postsynaptic expression of GluA1. Consistent with this, disrupting lipid rafts and GluA1 localization in rafts rescues Aβ-mediated suppression of hippocampal LTP. These findings unveil a novel functional deficit in GluA1 trafficking induced by Aβ, providing new insights into the mechanism underlying AD-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Ryosuke Midorikawa
- Department of Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yoshihiko Wakazono
- Department of Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
- Laboratory of Biophysical Research, Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Kogo Takamiya
- Department of Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
- Laboratory of Biophysical Research, Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan
| |
Collapse
|
14
|
de Vries LE, Huitinga I, Kessels HW, Swaab DF, Verhaagen J. The concept of resilience to Alzheimer's Disease: current definitions and cellular and molecular mechanisms. Mol Neurodegener 2024; 19:33. [PMID: 38589893 PMCID: PMC11003087 DOI: 10.1186/s13024-024-00719-7] [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: 08/23/2023] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Some individuals are able to maintain their cognitive abilities despite the presence of significant Alzheimer's Disease (AD) neuropathological changes. This discrepancy between cognition and pathology has been labeled as resilience and has evolved into a widely debated concept. External factors such as cognitive stimulation are associated with resilience to AD, but the exact cellular and molecular underpinnings are not completely understood. In this review, we discuss the current definitions used in the field, highlight the translational approaches used to investigate resilience to AD and summarize the underlying cellular and molecular substrates of resilience that have been derived from human and animal studies, which have received more and more attention in the last few years. From these studies the picture emerges that resilient individuals are different from AD patients in terms of specific pathological species and their cellular reaction to AD pathology, which possibly helps to maintain cognition up to a certain tipping point. Studying these rare resilient individuals can be of great importance as it could pave the way to novel therapeutic avenues for AD.
Collapse
Affiliation(s)
- Luuk E de Vries
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands.
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
| | - Helmut W Kessels
- Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Cabrera Y, Koymans KJ, Poe GR, Kessels HW, Van Someren EJW, Wassing R. Overnight neuronal plasticity and adaptation to emotional distress. Nat Rev Neurosci 2024; 25:253-271. [PMID: 38443627 DOI: 10.1038/s41583-024-00799-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: 02/01/2024] [Indexed: 03/07/2024]
Abstract
Expressions such as 'sleep on it' refer to the resolution of distressing experiences across a night of sound sleep. Sleep is an active state during which the brain reorganizes the synaptic connections that form memories. This Perspective proposes a model of how sleep modifies emotional memory traces. Sleep-dependent reorganization occurs through neurophysiological events in neurochemical contexts that determine the fates of synapses to grow, to survive or to be pruned. We discuss how low levels of acetylcholine during non-rapid eye movement sleep and low levels of noradrenaline during rapid eye movement sleep provide a unique window of opportunity for plasticity in neuronal representations of emotional memories that resolves the associated distress. We integrate sleep-facilitated adaptation over three levels: experience and behaviour, neuronal circuits, and synaptic events. The model generates testable hypotheses for how failed sleep-dependent adaptation to emotional distress is key to mental disorders, notably disorders of anxiety, depression and post-traumatic stress with the common aetiology of insomnia.
Collapse
Affiliation(s)
- Yesenia Cabrera
- Department of Integrative Biology and Physiology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Karin J Koymans
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Gina R Poe
- Department of Integrative Biology and Physiology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Helmut W Kessels
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
- Department of Synaptic Plasticity and Behaviour, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Society for Arts and Sciences, Amsterdam, Netherlands
| | - Eus J W Van Someren
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Society for Arts and Sciences, Amsterdam, Netherlands
- Department of Integrative Neurophysiology and Psychiatry, VU University, Amsterdam UMC, Amsterdam, Netherlands
- Center for Neurogenomics and Cognitive Research, VU University, Amsterdam UMC, Amsterdam, Netherlands
| | - Rick Wassing
- Sleep and Circadian Research, Woolcock Institute of Medical Research, Macquarie University, Sydney, New South Wales, Australia.
- School of Psychological Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia.
- Sydney Local Health District, Sydney, New South Wales, Australia.
| |
Collapse
|
16
|
Hao F, Bu Y, Huang S, Li W, Feng H, Wang Y. Maternal exposure to deltamethrin during pregnancy and lactation impairs neurodevelopment of male offspring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 274:116196. [PMID: 38461575 DOI: 10.1016/j.ecoenv.2024.116196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/28/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
Deltamethrin (DM) is a highly effective and widely used pyrethroid pesticide. It is an environmental factor affecting public and occupational health and exerts direct toxic effects on the central nervous system. As the major target organs for neurotoxicity of DM, the hippocampus and the cerebellum are critical to the learning and motor function. Pregnant Wistar rats were randomly divided into four groups and gavaged at doses of 0, 1, 4or 10 mg/kg/d DM from gestational day (GD) 0 to postnatal day (PN) 21. The PC12 cells were selected to further verify the regulatory mechanisms of DM on the neurodevelopmental injury. We found that maternal exposure to DM caused learning, memory and motor dysfunction in male offspring. Maternal exposure to DM induced the decrease in the density of hippocampal dendritic spines in male offspring through the reduced expression of M1 mAchRs, which in turn reduced the mediated AKT/mTOR signaling pathway, contributing to the inhibition of dynamic changes of GluA1. Meanwhile, DM exposure inhibited the BDNF/TrkB signaling pathway, thereby reducing phosphorylation of stathmin and impairing cerebellar purkinje cell dendrite growth and development. Taken together, maternal exposure to DM during pregnancy and lactation could impair neurodevelopment of male offspring.
Collapse
Affiliation(s)
- Fei Hao
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China; The Center for Disease Control and Prevention, Dalian Jinzhou New District, Dalian, China
| | - Ye Bu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China; Department of Planned Immunization, Liaoning Provincial Center for Disease Control and Prevention, Shenyang, China
| | - Shasha Huang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Wanqi Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Huiwen Feng
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Yuan Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China.
| |
Collapse
|
17
|
Araki Y, Rajkovich KE, Gerber EE, Gamache TR, Johnson RC, Tran THN, Liu B, Zhu Q, Hong I, Kirkwood A, Huganir R. SynGAP regulates synaptic plasticity and cognition independently of its catalytic activity. Science 2024; 383:eadk1291. [PMID: 38422154 PMCID: PMC11188940 DOI: 10.1126/science.adk1291] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/28/2023] [Indexed: 03/02/2024]
Abstract
SynGAP is an abundant synaptic GTPase-activating protein (GAP) critical for synaptic plasticity, learning, memory, and cognition. Mutations in SYNGAP1 in humans result in intellectual disability, autistic-like behaviors, and epilepsy. Heterozygous Syngap1-knockout mice display deficits in synaptic plasticity, learning, and memory and exhibit seizures. It is unclear whether SynGAP imparts structural properties at synapses independently of its GAP activity. Here, we report that inactivating mutations within the GAP domain do not inhibit synaptic plasticity or cause behavioral deficits. Instead, SynGAP modulates synaptic strength by physically competing with the AMPA-receptor-TARP excitatory receptor complex in the formation of molecular condensates with synaptic scaffolding proteins. These results have major implications for developing therapeutic treatments for SYNGAP1-related neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | | | | | - Richard C. Johnson
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thanh Hai N. Tran
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bian Liu
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qianwen Zhu
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alfredo Kirkwood
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
18
|
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
|
19
|
Liu QQ, Mi J, Du YY, Rong Z, Qin Y, Jiang W, Li X, Yu JY, Yang L, Du XY, Yang Q, Guo YY. Lotusine ameliorates propionic acid-induced autism spectrum disorder-like behavior in mice by activating D1 dopamine receptor in medial prefrontal cortex. Phytother Res 2024; 38:1089-1103. [PMID: 38168755 DOI: 10.1002/ptr.8098] [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: 06/15/2023] [Revised: 11/10/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
Autism spectrum disorder (ASD) is a multifaceted neuropsychiatric condition for which effective drug therapy for core clinical symptoms remains elusive. Lotusine, known for its neuroprotective properties in the treatment of neurological disorders, holds potential in addressing ASD. Nevertheless, its specific efficacy in ASD remains uncertain. This study aims to investigate the therapeutic potential of lotusine in ASD and elucidate the underlying molecular mechanisms. We induced an ASD mouse model through intracerebroventricular-propionic acid (ICV-PPA) injection for 7 days, followed by lotusine administration for 5 days. The efficacy of lotusine was evaluated through a battery of behavioral tests, including the three-chamber social test. The underlying mechanisms of lotusine action in ameliorating ASD-like behavior were investigated in the medial prefrontal cortex (mPFC) using whole-cell patch-clamp recordings, western blotting, immunofluorescence staining, molecular docking, and cellular thermal shift assay. The efficacy and mechanisms of lotusine were further validated in vitro. Lotusine effectively alleviated social deficits induced by ICV-PPA injection in mice by counteracting the reduction in miniature excitatory postsynaptic current frequency within the mPFC. Moreover, lotusine enhanced neuronal activity and ameliorated α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor dysfunction in ICV-PPA infusion mice by upregulating c-fos, p-GluA1 Ser 845, and p-GluA1 Ser 831 protein levels within the mPFC. Our findings also suggest that lotusine may exert its effects through modulation of the D1 dopamine receptor (DRD1). Furthermore, the rescuing effects of lotusine were nullified by a DRD1 antagonist in PC12 cells. In summary, our results revealed that lotusine ameliorates ASD-like behavior through targeted modulation of DRD1, ultimately enhancing excitatory synaptic transmission. These findings highlight the potential of lotusine as a nutritional supplement in the treatment of ASD.
Collapse
Affiliation(s)
- Qing-Qing Liu
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Jie Mi
- Department of Clinical Laboratory, Xi'an Children's Hospital, Xi'An, PR China
| | - Ya-Ya Du
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Zheng Rong
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Yan Qin
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Wei Jiang
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Xi Li
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Jiao-Yan Yu
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Le Yang
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Xiao-Yan Du
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Qi Yang
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| | - Yan-Yan Guo
- Department of Pharmacy, Tangdu Hospital, The Fourth Military Medical University, Xi'An, PR China
| |
Collapse
|
20
|
Tan JZA, Jang SE, Batallas-Borja A, Bhembre N, Chandra M, Zhang L, Guo H, Ringuet MT, Widagdo J, Collins BM, Anggono V. Copine-6 is a Ca 2+ sensor for activity-induced AMPA receptor exocytosis. Cell Rep 2023; 42:113460. [PMID: 37979168 DOI: 10.1016/j.celrep.2023.113460] [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: 04/13/2023] [Revised: 10/09/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
The recruitment of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors underlies the strengthening of neuronal connectivity during learning and memory. This process is triggered by N-methyl-D-aspartate (NMDA) receptor-dependent postsynaptic Ca2+ influx. Synaptotagmin (Syt)-1 and -7 have been proposed as Ca2+ sensors for AMPA receptor exocytosis but are functionally redundant. Here, we identify a cytosolic C2 domain-containing Ca2+-binding protein, Copine-6, that forms a complex with AMPA receptors. Loss of Copine-6 expression impairs activity-induced exocytosis of AMPA receptors in primary neurons, which is rescued by wild-type Copine-6 but not Ca2+-binding mutants. In contrast, Copine-6 loss of function does not affect steady-state expression or tetrodotoxin-induced synaptic upscaling of surface AMPA receptors. Loss of Syt-1/Syt-7 significantly reduces Copine-6 protein expression. Interestingly, overexpression of wild-type Copine-6, but not the Ca2+-binding mutants, restores activity-dependent exocytosis of AMPA receptors in Syt-1/Syt-7 double-knockdown neurons. We conclude that Copine-6 is a postsynaptic Ca2+ sensor that mediates AMPA receptor exocytosis during synaptic potentiation.
Collapse
Affiliation(s)
- Jing Zhi Anson Tan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Se Eun Jang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ana Batallas-Borja
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nishita Bhembre
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mintu Chandra
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Lingrui Zhang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Huimin Guo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mitchell T Ringuet
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
21
|
Chandravanshi LP, Agrawal P, Darwish HW, Trigun SK. Impairments of Spatial Memory and N-methyl-d-aspartate Receptors and Their Postsynaptic Signaling Molecules in the Hippocampus of Developing Rats Induced by As, Pb, and Mn Mixture Exposure. Brain Sci 2023; 13:1715. [PMID: 38137163 PMCID: PMC10742016 DOI: 10.3390/brainsci13121715] [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: 11/10/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Exposure to metal mixtures is recognized as a real-life scenario, needing novel studies that can assess their complex effects on brain development. There is still a significant public health concern associated with chronic low levels of metal exposure. In contrast to other metals, these three metals (As, Pb, and Mn) are commonly found in various environmental and industrial contexts. In addition to additive or synergistic interactions, concurrent exposure to this metal mixture may also have neurotoxic effects that differ from those caused by exposure to single components. The NMDA receptor and several important signaling proteins are involved in learning, memory, and synaptic plasticity in the hippocampus, including CaMKII, postsynaptic density protein-95 (PSD-95), synaptic Ras GTPase activating protein (SynGAP), a negative regulator of Ras-MAPK activity, and CREB. We hypothesized that alterations in the above molecular players may contribute to metal mixture developmental neurotoxicity. Thus, the aim of this study was to investigate the effect of these metals and their mixture at low doses (As 4 mg, Pb 4 mg, and Mn 10 mg/kg bw/p.o) on NMDA receptors and their postsynaptic signaling proteins during developing periods (GD6 to PD59) of the rat brain. Rats exposed to As, Pb, and Mn individually or at the same doses in a triple-metal mixture (MM) showed impairments in learning and memory functions in comparison to the control group rats. Declined protein expressions of NR2A, PSD-95, p- CaMKII, and pCREB were observed in the metal mix-exposed rats, while the expression of SynGAP was found to be enhanced in the hippocampus as compared to the controls on PD60. Thereby, our data suggest that alterations in the NMDA receptor complex and postsynaptic signaling proteins could explain the cognitive dysfunctions caused by metal-mixture-induced developmental neurotoxicity in rats. These outcomes indicate that incessant metal mixture exposure may have detrimental consequences on brain development.
Collapse
Affiliation(s)
- Lalit P. Chandravanshi
- Department of Forensic Science, Sharda University, Greater Noida 201308, India; (L.P.C.); (P.A.)
| | - Prashant Agrawal
- Department of Forensic Science, Sharda University, Greater Noida 201308, India; (L.P.C.); (P.A.)
| | - Hany W. Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Surendra Kumar Trigun
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
22
|
Tsugiyama LE, Macedo Moraes RC, Cavalcante Moraes YA, Francis-Oliveira J. Promising new pharmacological targets for depression: The search for efficacy. Drug Discov Today 2023; 28:103804. [PMID: 37865307 DOI: 10.1016/j.drudis.2023.103804] [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: 04/21/2023] [Revised: 08/31/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
Pharmacological treatment of major depressive disorder (MDD) still relies on the use of serotonergic drugs, despite their limited efficacy. A few mechanistically new drugs have been developed in recent years, but many fail in clinical trials. Several hypotheses have been proposed to explain MDD pathophysiology, indicating that physiological processes such as neuroplasticity, circadian rhythms, and metabolism are potential targets. Here, we review the current state of pharmacological treatments for MDD, as well as the preclinical and clinical evidence for an antidepressant effect of molecules that target non-serotonergic systems. We offer some insights into the challenges facing the development of new antidepressant drugs, and the prospect of finding more effectiveness for each target discussed.
Collapse
Affiliation(s)
- Lucila Emiko Tsugiyama
- Kansai Medical University, Graduate School of Medicine, iPS Cell Applied Medicine, Hirakata, Osaka, Japan
| | - Ruan Carlos Macedo Moraes
- University of Alabama at Birmingham, Department of Psychiatry and Behavioral Neurobiology, Birmingham, AL, USA; Biomedical Sciences Institute, Department of Human Physiology, Sao Paulo University, Sao Paulo, Brazil
| | | | - Jose Francis-Oliveira
- University of Alabama at Birmingham, Department of Psychiatry and Behavioral Neurobiology, Birmingham, AL, USA; Biomedical Sciences Institute, Department of Human Physiology, Sao Paulo University, Sao Paulo, Brazil.
| |
Collapse
|
23
|
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
|
24
|
Warming H, Deinhardt K, Garland P, More J, Bulters D, Galea I, Vargas-Caballero M. Functional effects of haemoglobin can be rescued by haptoglobin in an in vitro model of subarachnoid haemorrhage. J Neurochem 2023; 167:90-103. [PMID: 37702203 DOI: 10.1111/jnc.15936] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023]
Abstract
During subarachnoid haemorrhage, a blood clot forms in the subarachnoid space releasing extracellular haemoglobin (Hb), which causes oxidative damage and cell death in surrounding tissues. High rates of disability and cognitive decline in SAH survivors are attributed to loss of neurons and functional connections during secondary brain injury. Haptoglobin sequesters Hb for clearance, but this scavenging system is overwhelmed after a haemorrhage. Whilst exogenous haptoglobin application can attenuate cytotoxicity of Hb in vitro and in vivo, the functional effects of sub-lethal Hb concentrations on surviving neurons and whether cellular function can be protected with haptoglobin treatment remain unclear. Here we use cultured neurons to investigate neuronal health and function across a range of Hb concentrations to establish the thresholds for cellular damage and investigate synaptic function. Hb impairs ATP concentrations and cytoskeletal structure. At clinically relevant but sub-lethal Hb concentrations, we find that synaptic AMPAR-driven currents are reduced, accompanied by a reduction in GluA1 subunit expression. Haptoglobin co-application can prevent these deficits by scavenging free Hb to reduce it to sub-threshold concentrations and does not need to be present at stoichiometric amounts to achieve efficacy. Haptoglobin itself does not impair measures of neuronal health and function at any concentration tested. Our data highlight a role for Hb in modifying synaptic function in surviving neurons, which may link to impaired cognition or plasticity after SAH and support the development of haptoglobin as a therapy for subarachnoid haemorrhage.
Collapse
Affiliation(s)
- Hannah Warming
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Katrin Deinhardt
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | | | - John More
- Bio Products Laboratory Limited, Elstree, UK
| | - Diederik Bulters
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Mariana Vargas-Caballero
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
25
|
González-Alfonso WL, Pavel P, Karina HM, Del Razo LM, Sanchez-Peña LC, Zepeda A, Gonsebatt ME. Chronic exposure to inorganic arsenic and fluoride induces redox imbalance, inhibits the transsulfuration pathway, and alters glutamate receptor expression in the brain, resulting in memory impairment in adult male mouse offspring. Arch Toxicol 2023; 97:2371-2383. [PMID: 37482551 PMCID: PMC10404204 DOI: 10.1007/s00204-023-03556-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Exposure to toxic elements in drinking water, such as arsenic (As) and fluoride (F), starts at gestation and has been associated with memory and learning deficits in children. Studies in which rodents underwent mechanistic single exposure to As or F showed that the neurotoxic effects are associated with their capacity to disrupt redox balance, mainly by diminishing glutathione (GSH) levels, altering glutamate disposal, and altering glutamate receptor expression, which disrupts synaptic transmission. Elevated levels of As and F are common in groundwater worldwide. To explore the neurotoxicity of chronic exposure to As and F in drinking water, pregnant CD-1 mice were exposed to 2 mg/L As (sodium arsenite) and 25 mg/L F (sodium fluoride) alone or in combination. The male litter continued to receive exposure up to 30 or 90 days after birth. The effects of chronic exposure on GSH levels, transsulfuration pathway enzymatic activity, expression of cysteine/cystine transporters, glutamate transporters, and ionotropic glutamate receptor subunits as well as behavioral performance in the object recognition memory task were assessed. Combined exposure resulted in a significant reduction in GSH levels in the cortex and hippocampus at different times, decreased transsulfuration pathway enzyme activity, as well as diminished xCT protein expression. Altered glutamate receptor expression in the cortex and hippocampus and decreased transaminase enzyme activity were observed. These molecular alterations were associated with memory impairment in the object recognition task, which relies on these brain regions.
Collapse
Affiliation(s)
- Wendy L González-Alfonso
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México
| | - Petrosyan Pavel
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México
| | - Hernández-Mercado Karina
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México
| | - Luz M Del Razo
- Departamento de Toxicología, Centro de Investigación Y Estudios Avanzados, Mexico, DF, Mexico
| | - Luz C Sanchez-Peña
- Departamento de Toxicología, Centro de Investigación Y Estudios Avanzados, Mexico, DF, Mexico
| | - Angélica Zepeda
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México
| | - María E Gonsebatt
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, A. P. 70-228, Ciudad Universitaria, 04510, Mexico, CDMX, México.
| |
Collapse
|
26
|
Basli A, Bounaas J. Pathophysiological mechanism and natural preventive and therapeutic strategies of Alzheimer's disease. Nutr Health 2023; 29:403-413. [PMID: 36377316 DOI: 10.1177/02601060221137104] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) is characterized by the presence of two types of protein deposits in the brain, amyloid plaques and neurofibrillary tangles. The first one are dense deposits of beta amyloid protein, the second one are dense deposits of the protein tau. These proteins are present in all of our brains, but in AD they act unusually, leading to neuronal degeneration. This review will provide an overview of the AD, including the role of amyloid beta and tau, and mechanisms that lead to the formation of plaques and tangles. The review will also cover the existing researches that have focused on the inhibition of amyloid beta formation, cholinesterase, tau hyperphosphorylation, the pathogenic mechanisms of apoE4, and GSK-3 as a solution that could be used to slow or prevent the disease.
Collapse
Affiliation(s)
- Abdelkader Basli
- Laboratory of Interaction Research, Biodiversity, Ecosystems and Biotechnology, Faculty of Sciences, University of Skikda, Skikda, Algeria
| | - Jihane Bounaas
- Laboratory of Interaction Research, Biodiversity, Ecosystems and Biotechnology, Faculty of Sciences, University of Skikda, Skikda, Algeria
| |
Collapse
|
27
|
Zhang D, Ivica J, Krieger JM, Ho H, Yamashita K, Stockwell I, Baradaran R, Cais O, Greger IH. Structural mobility tunes signalling of the GluA1 AMPA glutamate receptor. Nature 2023; 621:877-882. [PMID: 37704721 PMCID: PMC10533411 DOI: 10.1038/s41586-023-06528-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 08/09/2023] [Indexed: 09/15/2023]
Abstract
AMPA glutamate receptors (AMPARs), the primary mediators of excitatory neurotransmission in the brain, are either GluA2 subunit-containing and thus Ca2+-impermeable, or GluA2-lacking and Ca2+-permeable1. Despite their prominent expression throughout interneurons and glia, their role in long-term potentiation and their involvement in a range of neuropathologies2, structural information for GluA2-lacking receptors is currently absent. Here we determine and characterize cryo-electron microscopy structures of the GluA1 homotetramer, fully occupied with TARPγ3 auxiliary subunits (GluA1/γ3). The gating core of both resting and open-state GluA1/γ3 closely resembles GluA2-containing receptors. However, the sequence-diverse N-terminal domains (NTDs) give rise to a highly mobile assembly, enabling domain swapping and subunit re-alignments in the ligand-binding domain tier that are pronounced in desensitized states. These transitions underlie the unique kinetic properties of GluA1. A GluA2 mutant (F231A) increasing NTD dynamics phenocopies this behaviour, and exhibits reduced synaptic responses, reflecting the anchoring function of the AMPAR NTD at the synapse. Together, this work underscores how the subunit-diverse NTDs determine subunit arrangement, gating properties and ultimately synaptic signalling efficiency among AMPAR subtypes.
Collapse
Affiliation(s)
- Danyang Zhang
- Neurobiology Division, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
| | - Josip Ivica
- Neurobiology Division, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
| | - James M Krieger
- Biocomputing Unit, National Center of Biotechnology, CSIC, Madrid, Spain
| | - Hinze Ho
- Neurobiology Division, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Keitaro Yamashita
- Structural Studies Division, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
| | - Imogen Stockwell
- Neurobiology Division, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
| | - Rozbeh Baradaran
- Neurobiology Division, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
| | - Ondrej Cais
- Neurobiology Division, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK
| | - Ingo H Greger
- Neurobiology Division, Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
28
|
Qneibi M, Jumaa H, Bdir S, Al-Maharik N. Electrophysiological Assessment of Newly Synthesized 2,3-Benzodiazepine Derivatives for Inhibiting the AMPA Receptor Channel. Molecules 2023; 28:6067. [PMID: 37630319 PMCID: PMC10458471 DOI: 10.3390/molecules28166067] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/08/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Three major subtypes of ionotropic receptors regulate glutamatergic synaptic transmission, one of which is α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (AMPARs). They are tetrameric, cation-permeable ionotropic glutamate receptors found across the brain. Abnormalities in AMPA receptor trafficking and synaptic assembly are linked to cognitive decline and neurological diseases such as Alzheimer's, Parkinson's, and Huntington's. The present study will investigate the effects of four novel 2,3-benzodiazepine derivatives on AMPA receptor subunits by comparing their effects on synaptic responses, desensitization, and deactivation rate in human embryonic kidney cells (HEK293T) recombinant AMPAR subunits using whole-cell patch-clamp electrophysiology. All four 2,3-BDZ compounds showed inhibitory activity against all the homomeric and heteromeric subunits tested. While the desensitization and deactivation rates in 2,3-BDZ-1 and 2,3-BDZ-2 decreased and increased, respectively, in the other two compounds (i.e., 2,3-BDZ-3 and 2,3-BDZ-4), there was no change in the desensitization or deactivation rates. These results contribute to a better understanding of AMPARs by identifying potential 2,3-BDZ drugs that demonstrate inhibitory effects on the AMPAR subunits.
Collapse
Affiliation(s)
- Mohammad Qneibi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine;
| | - Hanan Jumaa
- Department of Chemistry, Faculty of Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine;
| | - Sosana Bdir
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine;
| | - Nawaf Al-Maharik
- Department of Chemistry, Faculty of Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine;
| |
Collapse
|
29
|
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
|
30
|
Ireton KE, Xing X, Kim K, Weiner JC, Jacobi AA, Grover A, Foote M, Ota Y, Berman R, Hanks T, Hell JW. Regulation of the Ca 2+ Channel Ca V1.2 Supports Spatial Memory and Its Flexibility and LTD. J Neurosci 2023; 43:5559-5573. [PMID: 37419689 PMCID: PMC10376936 DOI: 10.1523/jneurosci.1521-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 04/30/2023] [Accepted: 05/15/2023] [Indexed: 07/09/2023] Open
Abstract
Widespread release of norepinephrine (NE) throughout the forebrain fosters learning and memory via adrenergic receptor (AR) signaling, but the molecular mechanisms are largely unknown. The β2 AR and its downstream effectors, the trimeric stimulatory Gs-protein, adenylyl cyclase (AC), and the cAMP-dependent protein kinase A (PKA), form a unique signaling complex with the L-type Ca2+ channel (LTCC) CaV1.2. Phosphorylation of CaV1.2 by PKA on Ser1928 is required for the upregulation of Ca2+ influx on β2 AR stimulation and long-term potentiation induced by prolonged theta-tetanus (PTT-LTP) but not LTP induced by two 1-s-long 100-Hz tetani. However, the function of Ser1928 phosphorylation in vivo is unknown. Here, we show that S1928A knock-in (KI) mice of both sexes, which lack PTT-LTP, express deficiencies during initial consolidation of spatial memory. Especially striking is the effect of this mutation on cognitive flexibility as tested by reversal learning. Mechanistically, long-term depression (LTD) has been implicated in reversal learning. It is abrogated in male and female S1928A knock-in mice and by β2 AR antagonists and peptides that displace β2 AR from CaV1.2. This work identifies CaV1.2 as a critical molecular locus that regulates synaptic plasticity, spatial memory and its reversal, and LTD.SIGNIFICANCE STATEMENT We show that phosphorylation of the Ca2+ channel CaV1.2 on Ser1928 is important for consolidation of spatial memory and especially its reversal, and long-term depression (LTD). Identification of Ser1928 as critical for LTD and reversal learning supports the model that LTD underlies flexibility of reference memory.
Collapse
Affiliation(s)
- Kyle E Ireton
- Department of Pharmacology, University of California, Davis, California 95616-8636
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Xiaoming Xing
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Karam Kim
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Justin C Weiner
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Ariel A Jacobi
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Aarushi Grover
- Department of Pharmacology, University of California, Davis, California 95616-8636
| | - Molly Foote
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Yusuke Ota
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Robert Berman
- Center for Neuroscience, University of California, Davis, California 95616-8636
| | - Timothy Hanks
- Center for Neuroscience, University of California, Davis, California 95616-8636
- Department of Neurology, University of California, Davis, California 95616-8636
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, California 95616-8636
- Center for Neuroscience, University of California, Davis, California 95616-8636
| |
Collapse
|
31
|
Lumeij LB, van Huijstee AN, Cappaert NLM, Kessels HW. Variance analysis as a method to predict the locus of plasticity at populations of non-uniform synapses. Front Cell Neurosci 2023; 17:1232541. [PMID: 37528963 PMCID: PMC10388551 DOI: 10.3389/fncel.2023.1232541] [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: 05/31/2023] [Accepted: 06/30/2023] [Indexed: 08/03/2023] Open
Abstract
Our knowledge on synaptic transmission in the central nervous system has often been obtained by evoking synaptic responses to populations of synapses. Analysis of the variance in synaptic responses can be applied as a method to predict whether a change in synaptic responses is a consequence of altered presynaptic neurotransmitter release or postsynaptic receptors. However, variance analysis is based on binomial statistics, which assumes that synapses are uniform. In reality, synapses are far from uniform, which questions the reliability of variance analysis when applying this method to populations of synapses. To address this, we used an in silico model for evoked synaptic responses and compared variance analysis outcomes between populations of uniform versus non-uniform synapses. This simulation revealed that variance analysis produces similar results irrespectively of the grade of uniformity of synapses. We put this variance analysis to the test with an electrophysiology experiment using a model system for which the loci of plasticity are well established: the effect of amyloid-β on synapses. Variance analysis correctly predicted that postsynaptically produced amyloid-β triggered predominantly a loss of synapses and a minor reduction of postsynaptic currents in remaining synapses with little effect on presynaptic release probability. We propose that variance analysis can be reliably used to predict the locus of synaptic changes for populations of non-uniform synapses.
Collapse
|
32
|
Furukawa M, Tada H, Raju R, Wang J, Yokoi H, Ikuyo Y, Yamada M, Shikama Y, Matsushita K. Long-Term Capsaicin Administration Ameliorates the Dysfunction and Astrogliosis of the Brain in Aged Mice with Missing Maxillary Molars. Nutrients 2023; 15:nu15112471. [PMID: 37299434 DOI: 10.3390/nu15112471] [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: 03/03/2023] [Revised: 05/13/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Tooth loss and decreased masticatory function reportedly affect cognitive function; tooth loss allegedly induces astrogliosis and aging of astrocytes in the hippocampus and hypothalamus, which is a response specific to the central nervous system owing to homeostasis in different brain regions. Capsaicin, a component of red peppers, has positive effects on brain disorders in mice. Decreased expression of transient receptor potential vanilloid 1, a receptor of capsaicin, is associated with the development of dementia. In this study, we investigated the effect of capsaicin administration in aged mice (C57BL/6N mice) with reduced masticatory function owing to the extraction of maxillary molars to investigate preventive/therapeutic methods for cognitive decline attributed to age-related masticatory function loss. The results demonstrated that mice with impaired masticatory function showed decreased motor and cognitive function at the behavioral level. At the genetic level, neuroinflammation, microglial activity, and astrogliosis, such as increased glial fibrillary acidic protein levels, were observed in the mouse brain. The mice with extracted molars fed on a diet containing capsaicin for 3 months demonstrated improved behavioral levels and astrogliosis, which suggest that capsaicin is useful in maintaining brain function in cases of poor oral function and prosthetic difficulties.
Collapse
Affiliation(s)
- Masae Furukawa
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Hirobumi Tada
- Department of Nutrition, Faculty of Wellness, Shigakkan University, Obu 474-8651, Japan
- Department of Integrative Physiology, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Resmi Raju
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Jingshu Wang
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Haruna Yokoi
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Yoriko Ikuyo
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Mitsuyoshi Yamada
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
- Department of Operative Dentistry, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan
| | - Yosuke Shikama
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Kenji Matsushita
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| |
Collapse
|
33
|
Qneibi M, Jaradat N, Al-Maharik N, Hawash M, Issa L, Suboh S, Yahya L, Khait AA, Warasneh A, Bdir S. The effect of Lavandula Coronopifolia essential oil on the biophysical properties of desensitization and deactivation gating currents in ionotropic receptors. Sci Rep 2023; 13:8417. [PMID: 37225859 DOI: 10.1038/s41598-023-35698-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 05/22/2023] [Indexed: 05/26/2023] Open
Abstract
The rising incidence of cancer and the lack of effective therapeutic interventions for many neurological illnesses like Alzheimer's and epilepsy has prompted us to investigate the composition and effects of the Lavandula coronopifolia oil from Palestine on cancer cells and AMPA receptor subunits in the brain due to the vast range of beneficial properties of Lavandula coronopifolia essential oil (EO). GC/MS was used to analyze L. coronopifolia's EO chemistry. EO's cytotoxicity and biophysical effects on AMPA receptors were investigated using MTS and electrophysiological techniques. The GC-MS results revealed that L. coronopifolia EO has a high content of eucalyptol (77.23%), β-pinene (6.93%), and α-pinene (4.95%). The EO showed more significant antiproliferative selectivity activities against HepG2 cancer cell lines than HEK293T cell lines with IC50 values of 58.51 and 133.22 µg/mL, respectively. The EO of L. coronopifolia affected AMPA receptor kinetics (desensitization and deactivation) and preferred homomeric GluA1 and heteromeric GluA1/A2 receptors. These findings indicate the potential therapeutic use of L. coronopifolia EO in the selective treatment of HepG2 cancer cell lines and neurodegenerative diseases.
Collapse
Affiliation(s)
- Mohammad Qneibi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine.
| | - Nawaf Al-Maharik
- Department of Chemistry, Faculty of Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Linda Issa
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Shorooq Suboh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Leen Yahya
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Adan Abu Khait
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Amjaad Warasneh
- Department of Pharmacy, 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
| |
Collapse
|
34
|
Català-Solsona J, Lituma PJ, Lutzu S, Siedlecki-Wullich D, Fábregas-Ordoñez C, Miñano-Molina AJ, Saura CA, Castillo PE, Rodriguez-Álvarez J. Activity-Dependent Nr4a2 Induction Modulates Synaptic Expression of AMPA Receptors and Plasticity via a Ca 2+/CRTC1/CREB Pathway. J Neurosci 2023; 43:3028-3041. [PMID: 36931707 PMCID: PMC10146469 DOI: 10.1523/jneurosci.1341-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
Transcription factors have a pivotal role in synaptic plasticity and the associated modification of neuronal networks required for memory formation and consolidation. The nuclear receptors subfamily 4 group A (Nr4a) have emerged as possible modulators of hippocampal synaptic plasticity and cognitive functions. However, the molecular and cellular mechanisms underlying Nr4a2-mediated hippocampal synaptic plasticity are not completely known. Here, we report that neuronal activity enhances Nr4a2 expression and function in cultured mouse hippocampal neurons (both sexes) by an ionotropic glutamate receptor/Ca2+/cAMP response element-binding protein/CREB-regulated transcription factor 1 (iGluR/Ca2+/CREB/CRTC1) pathway. Nr4a2 activation mediates BDNF production and increases expression of iGluRs, thereby affecting LTD at CA3-CA1 synapses in acute mouse hippocampal slices (both sexes). Together, our results indicate that the iGluR/Ca2+/CREB/CRTC1 pathway mediates activity-dependent expression of Nr4a2, which is involved in glutamatergic synaptic plasticity by increasing BDNF and synaptic GluA1-AMPARs. Therefore, Nr4a2 activation could be a therapeutic approach for brain disorders associated with dysregulated synaptic plasticity.SIGNIFICANCE STATEMENT A major factor that regulates fast excitatory synaptic transmission and plasticity is the modulation of synaptic AMPARs. However, despite decades of research, the underlying mechanisms of this modulation remain poorly understood. Our study identified a molecular pathway that links neuronal activity with AMPAR modulation and hippocampal synaptic plasticity through the activation of Nr4a2, a member of the nuclear receptor subfamily 4. Since several compounds have been described to activate Nr4a2, our study not only provides mechanistic insights into the molecular pathways related to hippocampal synaptic plasticity and learning, but also identifies Nr4a2 as a potential therapeutic target for pathologic conditions associated with dysregulation of glutamatergic synaptic function.
Collapse
Affiliation(s)
- Judit Català-Solsona
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Pablo J Lituma
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
| | - Stefano Lutzu
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
| | - Dolores Siedlecki-Wullich
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Cristina Fábregas-Ordoñez
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Alfredo J Miñano-Molina
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Carlos A Saura
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
- Department of Psychiatry & Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - José Rodriguez-Álvarez
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
| |
Collapse
|
35
|
Wang L, Tan Y, Wang H, Yu XD, Mo Y, Reilly J, He Z, Shu X. Urocanic acid facilitates acquisition of object recognition memory in mice. Physiol Behav 2023; 266:114201. [PMID: 37072048 DOI: 10.1016/j.physbeh.2023.114201] [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: 12/29/2022] [Revised: 03/13/2023] [Accepted: 04/14/2023] [Indexed: 04/20/2023]
Abstract
Trans-urocanic acid (UCA), an isomer of cis-UCA that is located in the skin, has recently been reported to have a role in short-term working memory and in the consolidation, reconsolidation and retrieval of long-term memory. However, its effect on memory acquisition remains unclear. In the present study, the effect of UCA on short-term and long-term memory acquisition in mice was investigated using novel object recognition (NOR) and object location recognition (OLR) protocols that each involved three stages: habituation, sampling and testing. UCA was intraperitoneally injected 0.5 h pre-sampling, and the discrimination index during subsequent testing was determined in NOR and OLR tasks. The results showed that 10 mg/kg UCA significantly facilitated short-term and long-term memory acquisition in both types of tasks. Furthermore, 30 mg/kg UCA significantly facilitated long-term memory acquisition in the NOR task and tended to facilitate long-term memory acquisition in the OLR tasks but did not facilitate short-term memory acquisition in either task. Additionally, the enhancing role of UCA on memory acquisition was not dependent on changes of nonspecific responses, e.g. exploratory behavior and locomotor activity. The current study suggests that UCA facilitates short-term and long-term recognition memory acquisition, which further extends the functional role of UCA in the brain function.
Collapse
Affiliation(s)
- Le Wang
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, P.R. China
| | - Yinna Tan
- Anesthesiology department, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421000, P.R
| | - Hao Wang
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, P.R. China
| | - Xu-Dong Yu
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, P.R. China
| | - Yanxin Mo
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, P.R. China
| | - James Reilly
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow G4 0BA, United Kingdom
| | - Zhiming He
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, P.R. China
| | - Xinhua Shu
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, P.R. China; Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow G4 0BA, United Kingdom; Department of Vision Science, Glasgow Caledonian University, Glasgow G4 0BA, United Kingdom.
| |
Collapse
|
36
|
Markiewicz-Gospodarek A, Markiewicz R, Borowski B, Dobrowolska B, Łoza B. Self-Regulatory Neuronal Mechanisms and Long-Term Challenges in Schizophrenia Treatment. Brain Sci 2023; 13:brainsci13040651. [PMID: 37190616 DOI: 10.3390/brainsci13040651] [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: 03/30/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Schizophrenia is a chronic and relapsing disorder that is characterized not only by delusions and hallucinations but also mainly by the progressive development of cognitive and social deficits. These deficits are related to impaired synaptic plasticity and impaired neurotransmission in the nervous system. Currently, technological innovations and medical advances make it possible to use various self-regulatory methods to improve impaired synaptic plasticity. To evaluate the therapeutic effect of various rehabilitation methods, we reviewed methods that modify synaptic plasticity and improve the cognitive and executive processes of patients with a diagnosis of schizophrenia. PubMed, Scopus, and Google Scholar bibliographic databases were searched with the keywords mentioned below. A total of 555 records were identified. Modern methods of schizophrenia therapy with neuroplastic potential, including neurofeedback, transcranial magnetic stimulation, transcranial direct current stimulation, vagus nerve stimulation, virtual reality therapy, and cognitive remediation therapy, were reviewed and analyzed. Since randomized controlled studies of long-term schizophrenia treatment do not exceed 2-3 years, and the pharmacological treatment itself has an incompletely estimated benefit-risk ratio, treatment methods based on other paradigms, including neuronal self-regulatory and neural plasticity mechanisms, should be considered. Methods available for monitoring neuroplastic effects in vivo (e.g., fMRI, neuropeptides in serum), as well as unfavorable parameters (e.g., features of the metabolic syndrome), enable individualized monitoring of the effectiveness of long-term treatment of schizophrenia.
Collapse
Affiliation(s)
| | - Renata Markiewicz
- Department of Neurology, Neurological and Psychiatric Nursing, Medical University of Lublin, 20-093 Lublin, Poland
| | - Bartosz Borowski
- Students Scientific Association at the Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Beata Dobrowolska
- Department of Holistic Care and Management in Nursing, Medical University of Lublin, 20-081 Lublin, Poland
| | - Bartosz Łoza
- Department of Psychiatry, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
37
|
Eiro T, Miyazaki T, Hatano M, Nakajima W, Arisawa T, Takada Y, Kimura K, Sano A, Nakano K, Mihara T, Takayama Y, Ikegaya N, Iwasaki M, Hishimoto A, Noda Y, Miyazaki T, Uchida H, Tani H, Nagai N, Koizumi T, Nakajima S, Mimura M, Matsuda N, Kanai K, Takahashi K, Ito H, Hirano Y, Kimura Y, Matsumoto R, Ikeda A, Takahashi T. Dynamics of AMPA receptors regulate epileptogenesis in patients with epilepsy. Cell Rep Med 2023; 4:101020. [PMID: 37080205 DOI: 10.1016/j.xcrm.2023.101020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/08/2023] [Accepted: 03/22/2023] [Indexed: 04/22/2023]
Abstract
The excitatory glutamate α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) contribute to epileptogenesis. Thirty patients with epilepsy and 31 healthy controls are scanned using positron emission tomography with our recently developed radiotracer for AMPARs, [11C]K-2, which measures the density of cell-surface AMPARs. In patients with focal-onset seizures, an increase in AMPAR trafficking augments the amplitude of abnormal gamma activity detected by electroencephalography. In contrast, patients with generalized-onset seizures exhibit a decrease in AMPARs coupled with increased amplitude of abnormal gamma activity. Patients with epilepsy had reduced AMPAR levels compared with healthy controls, and AMPARs are reduced in larger areas of the cortex in patients with generalized-onset seizures compared with those with focal-onset seizures. Thus, epileptic brain function can be regulated by the enhanced trafficking of AMPAR due to Hebbian plasticity with increased simultaneous neuronal firing and compensational downregulation of cell-surface AMPARs by the synaptic scaling.
Collapse
Affiliation(s)
- Tsuyoshi Eiro
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; Department of Psychiatry, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Tomoyuki Miyazaki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Mai Hatano
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Waki Nakajima
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Tetsu Arisawa
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Yuuki Takada
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kimito Kimura
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Akane Sano
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kotaro Nakano
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Takahiro Mihara
- Department of Health Data Science, Yokohama City University Graduate School of Data Science, Yokohama 236-0004, Japan
| | - Yutaro Takayama
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Naoki Ikegaya
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Masaki Iwasaki
- Department of Neurosurgery, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira 187-8551, Japan
| | - Akitoyo Hishimoto
- Department of Psychiatry, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Takahiro Miyazaki
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Hideaki Tani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Nobuhiro Nagai
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Teruki Koizumi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Nozomu Matsuda
- Department of Neurology, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Kazuaki Kanai
- Department of Neurology, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hiroshi Ito
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima 960-1295, Japan; Department of Radiology and Nuclear Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Yoji Hirano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Department of Psychiatry, Division of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Yuichi Kimura
- Faculty of Informatics, Cyber Informatics Research Institute, Kindai University, Higashi-Osaka 577-8502, Japan
| | - Riki Matsumoto
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Akio Ikeda
- Department of Epilepsy, Movement Disorders and Physiology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan; The University of Tokyo, International Research Center for Neurointelligence, Tokyo 113-0033, Japan.
| |
Collapse
|
38
|
Zhang J, Liu Z, Liu X, Wang X, Yu L. Intravenous Injection of GluR2-3Y Inhibits Repeated Morphine-Primed Reinstatement of Drug Seeking in Rats. Brain Sci 2023; 13:brainsci13040590. [PMID: 37190555 DOI: 10.3390/brainsci13040590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Studies have demonstrated that the α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor is essential to drug addiction. In this study, we explored the influence of GluR2-3Y, an interfering peptide to prevent the endocytosis of AMPA receptors containing the GluR2 subunit, on morphine-seeking behavior in the rat self-administration model. After self-administration was established, the rats received intravenous injections of GluR2-3Y during the extinction sessions. There were no significant differences in both active and inactive pokes compared to the control group of rats that received GluR2-3S, indicating that GluR2-3Y has no significant influences on the extinction of morphine self-administration. The other two groups of rats were trained, extinguished, and reinstated by repeated morphine priming (respectively, called Prime 1, Prime 2, and Prime 3). Only one intravenous injection of GluR2-3Y was performed before Prime 1. Compared to the control group, GluR2-3Y did not affect Prime 1, but significantly attenuated the morphine-seeking behavior during repeated morphine-primed reinstatement, indicating an inhibitory after effect of GluR2-3Y on morphine-seeking behavior in rats. The long-term depression (LTD) in the nucleus accumbens (NAc) shell was also assessed. Pretreatment with GluR2-3Y altered the ability of LTD induction to the level of that in the naive group, while pretreatment with GluR2-3S had no effects on LTD. Our results demonstrated that the intravenous injection of GluR2-3Y, to block the endocytosis of AMPA receptors, inhibited the reinstatement of morphine-seeking behavior, which may be induced by modulating the neuronal plasticity in the NAc shell of rats.
Collapse
Affiliation(s)
- Jianjun Zhang
- College of Basic Medical, Shanxi University of Chinese Medicine, Jinzhong 030619, China
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong 030619, China
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing 100101, China
| | - Zhuo Liu
- School of Crime Investigation, People’s Public Security University of China, Beijing 100038, China
| | - Xiaodong Liu
- Beijing Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoqian Wang
- College of Basic Medical, Shanxi University of Chinese Medicine, Jinzhong 030619, China
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong 030619, China
| | - Longchuan Yu
- School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
39
|
Jeong S, Kang HW, Kim SH, Hong GS, Nam MH, Seong J, Yoon ES, Cho IJ, Chung S, Bang S, Kim HN, Choi N. Integration of reconfigurable microchannels into aligned three-dimensional neural networks for spatially controllable neuromodulation. SCIENCE ADVANCES 2023; 9:eadf0925. [PMID: 36897938 PMCID: PMC10005277 DOI: 10.1126/sciadv.adf0925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Anisotropically organized neural networks are indispensable routes for functional connectivity in the brain, which remains largely unknown. While prevailing animal models require additional preparation and stimulation-applying devices and have exhibited limited capabilities regarding localized stimulation, no in vitro platform exists that permits spatiotemporal control of chemo-stimulation in anisotropic three-dimensional (3D) neural networks. We present the integration of microchannels seamlessly into a fibril-aligned 3D scaffold by adapting a single fabrication principle. We investigated the underlying physics of elastic microchannels' ridges and interfacial sol-gel transition of collagen under compression to determine a critical window of geometry and strain. We demonstrated the spatiotemporally resolved neuromodulation in an aligned 3D neural network by local deliveries of KCl and Ca2+ signal inhibitors, such as tetrodotoxin, nifedipine, and mibefradil, and also visualized Ca2+ signal propagation with a speed of ~3.7 μm/s. We anticipate that our technology will pave the way to elucidate functional connectivity and neurological diseases associated with transsynaptic propagation.
Collapse
Affiliation(s)
- Sohyeon Jeong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- MEPSGEN Co. Ltd., Seoul 05836, Korea
| | - Hyun Wook Kang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- School of Mechanical Engineering, Korea University, Seoul 02841, Korea
| | - So Hyun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- SK Biopharmaceuticals Co. Ltd., Seongnam 13494, Korea
| | - Gyu-Sang Hong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- Department of Life Sciences, Korea University, Seoul 02841, Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02453, Korea
| | - Eui-Sung Yoon
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Nano and Information Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
| | - Il-Joo Cho
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
| | - Seok Chung
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- School of Mechanical Engineering, Korea University, Seoul 02841, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Department of Medical Biotechnology, Dongguk University, Goyang 10326, Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| |
Collapse
|
40
|
Naylor DE. In the fast lane: Receptor trafficking during status epilepticus. Epilepsia Open 2023; 8 Suppl 1:S35-S65. [PMID: 36861477 PMCID: PMC10173858 DOI: 10.1002/epi4.12718] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Status epilepticus (SE) remains a significant cause of morbidity and mortality and often is refractory to standard first-line treatments. A rapid loss of synaptic inhibition and development of pharmacoresistance to benzodiazepines (BZDs) occurs early during SE, while NMDA and AMPA receptor antagonists remain effective treatments after BZDs have failed. Multimodal and subunit-selective receptor trafficking within minutes to an hour of SE involves GABA-A, NMDA, and AMPA receptors and contributes to shifts in the number and subunit composition of surface receptors with differential impacts on the physiology, pharmacology, and strength of GABAergic and glutamatergic currents at synaptic and extrasynaptic sites. During the first hour of SE, synaptic GABA-A receptors containing γ2 subunits move to the cell interior while extrasynaptic GABA-A receptors with δ subunits are preserved. Conversely, NMDA receptors containing N2B subunits are increased at synaptic and extrasynaptic sites, and homomeric GluA1 ("GluA2-lacking") calcium permeant AMPA receptor surface expression also is increased. Molecular mechanisms, largely driven by NMDA receptor or calcium permeant AMPA receptor activation early during circuit hyperactivity, regulate subunit-specific interactions with proteins involved with synaptic scaffolding, adaptin-AP2/clathrin-dependent endocytosis, endoplasmic reticulum (ER) retention, and endosomal recycling. Reviewed here is how SE-induced shifts in receptor subunit composition and surface representation increase the excitatory to inhibitory imbalance that sustains seizures and fuels excitotoxicity contributing to chronic sequela such as "spontaneous recurrent seizures" (SRS). A role for early multimodal therapy is suggested both for treatment of SE and for prevention of long-term comorbidities.
Collapse
Affiliation(s)
- David E Naylor
- VA Greater Los Angeles Healthcare System, Department of Neurology, David Geffen School of Medicine at UCLA, and The Lundquist Institute at Harbor-UCLA Medical Center, Los Angeles, California, USA
| |
Collapse
|
41
|
Mulvey B, Frye HE, Lintz T, Fass S, Tycksen E, Nelson EC, Morón JA, Dougherty JD. Cnih3 Deletion Dysregulates Dorsal Hippocampal Transcription across the Estrous Cycle. eNeuro 2023; 10:ENEURO.0153-22.2023. [PMID: 36849260 PMCID: PMC10027183 DOI: 10.1523/eneuro.0153-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 03/01/2023] Open
Abstract
In females, the hippocampus, a critical brain region for coordination of learning, memory, and behavior, displays altered physiology and behavioral output across the estrous or menstrual cycle. However, the molecular effectors and cell types underlying these observed cyclic changes have only been partially characterized to date. Recently, profiling of mice null for the AMPA receptor trafficking gene Cnih3 have demonstrated estrous-dependent phenotypes in dorsal hippocampal synaptic plasticity, composition, and learning/memory. We therefore profiled dorsal hippocampal transcriptomes from female mice in each estrous cycle stage, and contrasted it with that of males, across wild-type (WT) and Cnih3 mutants. In wild types, we identified only subtle differences in gene expression between the sexes, while comparing estrous stages to one another revealed up to >1000 differentially expressed genes (DEGs). These estrous-responsive genes are especially enriched in gene markers of oligodendrocytes and the dentate gyrus, and in functional gene sets relating to estrogen response, potassium channels, and synaptic gene splicing. Surprisingly, Cnih3 knock-outs (KOs) showed far broader transcriptomic differences between estrous cycle stages and males. Moreover, Cnih3 knock-out drove subtle but extensive expression changes accentuating sex differential expression at diestrus and estrus. Altogether, our profiling highlights cell types and molecular systems potentially impacted by estrous-specific gene expression patterns in the adult dorsal hippocampus, enabling mechanistic hypothesis generation for future studies of sex-differential neuropsychiatric function and dysfunction. Moreover, these findings suggest an unrecognized role of Cnih3 in buffering against transcriptional effects of estrous, providing a candidate molecular mechanism to explain estrous-dependent phenotypes observed with Cnih3 loss.
Collapse
Affiliation(s)
- Bernard Mulvey
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Hannah E Frye
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Tania Lintz
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Stuart Fass
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Eric Tycksen
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110
| | - Elliot C Nelson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Jose A Morón
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
42
|
Chakraborty P, Dey A, Gopalakrishnan AV, Swati K, Ojha S, Prakash A, Kumar D, Ambasta RK, Jha NK, Jha SK, Dewanjee S. Glutamatergic neurotransmission: A potential pharmacotherapeutic target for the treatment of cognitive disorders. Ageing Res Rev 2023; 85:101838. [PMID: 36610558 DOI: 10.1016/j.arr.2022.101838] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023]
Abstract
In the mammalian brain, glutamate is regarded to be the primary excitatory neurotransmitter due to its widespread distribution and wide range of metabolic functions. Glutamate plays key roles in regulating neurogenesis, synaptogenesis, neurite outgrowth, and neuron survival in the brain. Ionotropic and metabotropic glutamate receptors, neurotransmitters, neurotensin, neurosteroids, and others co-ordinately formulate a complex glutamatergic network in the brain that maintains optimal excitatory neurotransmission. Cognitive activities are potentially synchronized by the glutamatergic activities in the brain via restoring synaptic plasticity. Dysfunctional glutamate receptors and other glutamatergic components are responsible for the aberrant glutamatergic activity in the brain that cause cognitive impairments, loss of synaptic plasticity, and neuronal damage. Thus, controlling the brain's glutamatergic transmission and modifying glutamate receptor function could be a potential therapeutic strategy for cognitive disorders. Certain drugs that regulate glutamate receptor activities have shown therapeutic promise in improving cognitive functions in preclinical and clinical studies. However, several issues regarding precise functional information of glutamatergic activity are yet to be comprehensively understood. The present article discusses the scope of developing glutamatergic systems as prospective pharmacotherapeutic targets to treat cognitive disorders. Special attention has been given to recent developments, challenges, and future prospects.
Collapse
Affiliation(s)
- Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, West Bengal, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Kumari Swati
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Anand Prakash
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand 248007, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India.
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
43
|
Rollenhagen A, Anstötz M, Zimmermann K, Kasugai Y, Sätzler K, Molnar E, Ferraguti F, Lübke JHR. Layer-specific distribution and expression pattern of AMPA- and NMDA-type glutamate receptors in the barrel field of the adult rat somatosensory cortex: a quantitative electron microscopic analysis. Cereb Cortex 2023; 33:2342-2360. [PMID: 35732315 PMCID: PMC9977369 DOI: 10.1093/cercor/bhac212] [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: 12/09/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/14/2022] Open
Abstract
AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) and NMDA (N-methyl-d-aspartate) glutamate receptors are driving forces for synaptic transmission and plasticity at neocortical synapses. However, their distribution pattern in the adult rat neocortex is largely unknown and was quantified using freeze fracture replication combined with postimmunogold-labeling. Both receptors were co-localized at layer (L)4 and L5 postsynaptic densities (PSDs). At L4 dendritic shaft and spine PSDs, the number of gold grains detecting AMPA was similar, whereas at L5 shaft PSDs AMPA-receptors outnumbered those on spine PSDs. Their number was significantly higher at L5 vs. L4 PSDs. At L4 and L5 dendritic shaft PSDs, the number of gold grains detecting GluN1 was ~2-fold higher than at spine PSDs. The number of gold grains detecting the GluN1-subunit was higher for both shaft and spine PSDs in L5 vs. L4. Both receptors showed a large variability in L4 and L5. A high correlation between the number of gold grains and PSD size for both receptors and targets was observed. Both receptors were distributed over the entire PSD but showed a layer- and target-specific distribution pattern. The layer- and target-specific distribution of AMPA and GluN1 glutamate receptors partially contribute to the observed functional differences in synaptic transmission and plasticity in the neocortex.
Collapse
Affiliation(s)
- Astrid Rollenhagen
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany
| | - Max Anstötz
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany.,Institute of Anatomy II, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Universitätsstr. 1, Düsseldorf 40001, Germany
| | - Kerstin Zimmermann
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany
| | - Yu Kasugai
- Department of Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1a, Innsbruck A-6020, Austria
| | - Kurt Sätzler
- School of Biomedical Sciences, University of Ulster, Cromore Rd., Londonderry BT52 1SA, United Kingdom
| | - Elek Molnar
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1a, Innsbruck A-6020, Austria
| | - Joachim H R Lübke
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH/Medical University Aachen, Pauwelstr. 30, Aachen 52074, Germany.,JARA Translational Medicine Jülich/Aachen, Germany
| |
Collapse
|
44
|
Hussein A, Guevara CA, Valle PD, Gupta S, Benson DL, Huntley GW. Non-Motor Symptoms of Parkinson's Disease: The Neurobiology of Early Psychiatric and Cognitive Dysfunction. Neuroscientist 2023; 29:97-116. [PMID: 33966533 PMCID: PMC9338765 DOI: 10.1177/10738584211011979] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that has been recognized for over 200 years by its clinically dominant motor system impairment. There are prominent non-motor symptoms as well, and among these, psychiatric symptoms of depression and anxiety and cognitive impairment are common and can appear earlier than motor symptoms. Although the neurobiology underlying these particular PD-associated non-motor symptoms is not completely understood, the identification of PARK genes that contribute to hereditary and sporadic PD has enabled genetic models in animals that, in turn, have fostered ever deepening analyses of cells, synapses, circuits, and behaviors relevant to non-motor psychiatric and cognitive symptoms of human PD. Moreover, while it has long been recognized that inflammation is a prominent component of PD, recent studies demonstrate that brain-immune signaling crosstalk has significant modulatory effects on brain cell and synaptic function in the context of psychiatric symptoms. This review provides a focused update on such progress in understanding the neurobiology of PD-related non-motor psychiatric and cognitive symptoms.
Collapse
Affiliation(s)
- Ayan Hussein
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher A. Guevara
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pamela Del Valle
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Swati Gupta
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deanna L. Benson
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - George W. Huntley
- Nash Family Department of Neuroscience and Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
45
|
Schwarz K, Schmitz F. Synapse Dysfunctions in Multiple Sclerosis. Int J Mol Sci 2023; 24:ijms24021639. [PMID: 36675155 PMCID: PMC9862173 DOI: 10.3390/ijms24021639] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neuroinflammatory disease of the central nervous system (CNS) affecting nearly three million humans worldwide. In MS, cells of an auto-reactive immune system invade the brain and cause neuroinflammation. Neuroinflammation triggers a complex, multi-faceted harmful process not only in the white matter but also in the grey matter of the brain. In the grey matter, neuroinflammation causes synapse dysfunctions. Synapse dysfunctions in MS occur early and independent from white matter demyelination and are likely correlates of cognitive and mental symptoms in MS. Disturbed synapse/glia interactions and elevated neuroinflammatory signals play a central role. Glutamatergic excitotoxic synapse damage emerges as a major mechanism. We review synapse/glia communication under normal conditions and summarize how this communication becomes malfunctional during neuroinflammation in MS. We discuss mechanisms of how disturbed glia/synapse communication can lead to synapse dysfunctions, signaling dysbalance, and neurodegeneration in MS.
Collapse
|
46
|
Li JB, Hu XY, Chen MW, Xiong CH, Zhao N, Ge YH, Wang H, Gao XL, Xu NJ, Zhao LX, Yu ZH, Chen HZ, Qiu Y. p85S6K sustains synaptic GluA1 to ameliorate cognitive deficits in Alzheimer's disease. Transl Neurodegener 2023; 12:1. [PMID: 36624510 PMCID: PMC9827685 DOI: 10.1186/s40035-022-00334-w] [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: 07/03/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Ribosomal protein S6 kinase 1 (S6K1) is a serine-threonine kinase that has two main isoforms: p70S6K (70-kDa isoform) and p85S6K (85-kDa isoform). p70S6K, with its upstream mammalian target of rapamycin (mTOR), has been shown to be involved in learning and memory and participate in the pathophysiology of Alzheimer's disease (AD). However, the function of p85S6K has long been neglected due to its high similarity to p70S6k. The role of p85S6K in learning and memory is still largely unknown. METHODS We fractionated the postsynaptic densities to illustrate the differential distribution of p85S6K and p70S6K. Coimmunoprecipitation was performed to unveil interactions between p85S6K and the GluA1 subunit of AMPA receptor. The roles of p85S6K in synaptic targeting of GluA1 and learning and memory were evaluated by specific knockdown or overexpression of p85S6K followed by a broad range of methodologies including immunofluorescence, Western blot, in situ proximity ligation assay, morphological staining and behavioral examination. Further, the expression level of p85S6K was measured in brains from AD patients and AD model mice. RESULTS p85S6K, but not p70S6K, was enriched in the postsynaptic densities. Moreover, knockdown of p85S6K resulted in defective spatial and recognition memory. In addition, p85S6K could interact with the GluA1 subunit of AMPA receptor through synapse-associated protein 97 and A-kinase anchoring protein 79/150. Mechanistic studies demonstrated that p85S6K could directly phosphorylate GluA1 at Ser845 and increase the amount of GluA1 in synapses, thus sustaining synaptic function and spine densities. Moreover, p85S6K was found to be specifically decreased in the synaptosomal compartment in the brains of AD patients and AD mice. Overexpression of p85S6K ameliorated the synaptic deficits and cognitive impairment in transgenic AD model mice. CONCLUSIONS These results strongly imply a significant role for p85S6K in maintaining synaptic and cognitive function by interacting with GluA1. The findings provide an insight into the rational targeting of p85S6K as a therapeutic potential for AD.
Collapse
Affiliation(s)
- Jia-Bing Li
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xiao-Yu Hu
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Mu-Wen Chen
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Cai-Hong Xiong
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Na Zhao
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Yan-Hui Ge
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Hao Wang
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xiao-Ling Gao
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Nan-Jie Xu
- grid.16821.3c0000 0004 0368 8293Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Lan-Xue Zhao
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Zhi-Hua Yu
- grid.16821.3c0000 0004 0368 8293Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Yu Qiu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
47
|
Nieves Torres D, Lee SH. Inter-neuronal signaling mediated by small extracellular vesicles: wireless communication? Front Mol Neurosci 2023; 16:1187300. [PMID: 37181650 PMCID: PMC10172472 DOI: 10.3389/fnmol.2023.1187300] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/03/2023] [Indexed: 05/16/2023] Open
Abstract
Conventional inter-neuronal communication conceptualizes the wired method of chemical synapses that physically connect pre-and post-synaptic neurons. In contrast, recent studies indicate that neurons also utilize synapse-independent, hence "wireless" broadcasting-type communications via small extracellular vesicles (EVs). Small EVs including exosomes are secreted vesicles released by cells and contain a variety of signaling molecules including mRNAs, miRNAs, lipids, and proteins. Small EVs are subsequently absorbed by local recipient cells via either membrane fusion or endocytic processes. Therefore, small EVs enable cells to exchange a "packet" of active biomolecules for communication purposes. It is now well established that central neurons also secrete and uptake small EVs, especially exosomes, a type of small EVs that are derived from the intraluminal vesicles of multivesicular bodies. Specific molecules carried by neuronal small EVs are shown to affect a variety of neuronal functions including axon guidance, synapse formation, synapse elimination, neuronal firing, and potentiation. Therefore, this type of volume transmission mediated by small EVs is thought to play important roles not only in activity-dependent changes in neuronal function but also in the maintenance and homeostatic control of local circuitry. In this review, we summarize recent discoveries, catalog neuronal small EV-specific biomolecules, and discuss the potential scope of small EV-mediated inter-neuronal signaling.
Collapse
Affiliation(s)
- Damaris Nieves Torres
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sang H Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
- Neuroscience Research Institute, Medical College of Wisconsin, Milwaukee, WI, United States
- *Correspondence: Sang H. Lee,
| |
Collapse
|
48
|
Demin KA, Krotova NA, Ilyin NP, Galstyan DS, Kolesnikova TO, Strekalova T, de Abreu MS, Petersen EV, Zabegalov KN, Kalueff AV. Evolutionarily conserved gene expression patterns for affective disorders revealed using cross-species brain transcriptomic analyses in humans, rats and zebrafish. Sci Rep 2022; 12:20836. [PMID: 36460699 PMCID: PMC9718822 DOI: 10.1038/s41598-022-22688-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
Widespread, debilitating and often treatment-resistant, depression and other stress-related neuropsychiatric disorders represent an urgent unmet biomedical and societal problem. Although animal models of these disorders are commonly used to study stress pathogenesis, they are often difficult to translate across species into valuable and meaningful clinically relevant data. To address this problem, here we utilized several cross-species/cross-taxon approaches to identify potential evolutionarily conserved differentially expressed genes and their sets. We also assessed enrichment of these genes for transcription factors DNA-binding sites down- and up- stream from their genetic sequences. For this, we compared our own RNA-seq brain transcriptomic data obtained from chronically stressed rats and zebrafish with publicly available human transcriptomic data for patients with major depression and their respective healthy control groups. Utilizing these data from the three species, we next analyzed their differential gene expression, gene set enrichment and protein-protein interaction networks, combined with validated tools for data pooling. This approach allowed us to identify several key brain proteins (GRIA1, DLG1, CDH1, THRB, PLCG2, NGEF, IKZF1 and FEZF2) as promising, evolutionarily conserved and shared affective 'hub' protein targets, as well as to propose a novel gene set that may be used to further study affective pathogenesis. Overall, these approaches may advance cross-species brain transcriptomic analyses, and call for further cross-species studies into putative shared molecular mechanisms of affective pathogenesis.
Collapse
Affiliation(s)
- Konstantin A Demin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
| | - Nataliya A Krotova
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Nikita P Ilyin
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | | | | | | | | | | | - Allan V Kalueff
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia.
- Institute of Neurosciences and Medicine, Novosibirsk, Russia.
- Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
49
|
Gómez de San José N, Goossens J, Al Shweiki MR, Halbgebauer S, Oeckl P, Steinacker P, Danzer KM, Graf H, Schönfeldt-Lecuona C, Belbin O, Lleó A, Vanmechelen E, Otto M. Glutamate receptor 4 as a fluid biomarker for the diagnosis of psychiatric disorders. J Psychiatr Res 2022; 156:390-397. [PMID: 36323141 DOI: 10.1016/j.jpsychires.2022.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/02/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
Psychiatric disorders are widely underreported diseases, especially in their early stages. So far, there is no fluid biomarker to confirm the diagnosis of these disorders. Proteomics data suggest the synaptic protein glutamate receptor 4 (GluR4), part of the AMPA receptor, as a potential diagnostic biomarker of major depressive disorder (MDD). A novel sandwich ELISA was established and analytically validated to detect GluR4 in cerebrospinal fluid (CSF) samples. A total of 85 subjects diagnosed with MDD (n = 36), bipolar disorder (BD, n = 12), schizophrenia (SCZ, n = 12) and neurological controls (CON, n = 25) were analysed. The data exhibited a significant correlation (r = 0.74; CI:0.62 to 0.82; p < 0.0001) with the antibody-free multiple reaction monitoring (MRM) mass spectrometry (MS) data. CSF GluR4 levels were lower in MDD (p < 0.002) and BD (p = 0.012) than in CON. Moreover, subjects with SCZ described a trend towards lower levels than CON (p = 0.13). The novel GluR4 ELISA may favour the clinical application of this protein as a potential diagnostic biomarker of psychiatric disorders and may facilitate the understanding of the pathophysiological mechanisms behind these disorders.
Collapse
Affiliation(s)
| | | | | | - Steffen Halbgebauer
- Department of Neurology, University of Ulm, 89075, Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE E.V.), Ulm, Germany.
| | - Patrick Oeckl
- Department of Neurology, University of Ulm, 89075, Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE E.V.), Ulm, Germany.
| | - Petra Steinacker
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120, Halle (Saale), Germany.
| | - Karin M Danzer
- Department of Neurology, University of Ulm, 89075, Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE E.V.), Ulm, Germany.
| | - Heiko Graf
- Department of Psychiatry and Psychotherapy III, University of Ulm, 89075, Ulm, Germany.
| | | | - Olivia Belbin
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
| | - Alberto Lleó
- Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Sant Antoni Maria Claret, 167, 08025, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
| | | | - Markus Otto
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120, Halle (Saale), Germany.
| |
Collapse
|
50
|
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: 11] [Impact Index Per Article: 5.5] [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
|