1
|
Kim KW, Lee E, Ko A, Hwang J, Park K, Lee BC, Kim KW, Oh WJ, Kim K, Lim HH. Cryo-EM structures of mouse bestrophin 1 channel in closed and partially open conformations. Mol Cells 2025; 48:100208. [PMID: 40043778 PMCID: PMC11964753 DOI: 10.1016/j.mocell.2025.100208] [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: 12/08/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
Bestrophin 1 (BEST1) channels are calcium-activated Cl- channels involved in diverse physiological processes, including gliotransmitter release in astrocytes. Although human and chicken BEST1 orthologs have been extensively studied, the structural and functional properties of mouse BEST1 (mBEST1) remain poorly understood. In this study, we characterized the structure-function of mBEST1-BF, a C-terminally tagged variant, using whole-cell patch-clamp recordings, surface biotinylation assays, and single-particle cryo-electron microscopy. Cryo-electron microscopy structural analysis of mBEST1-BF revealed closed and partially open conformations. Comparative analysis with human and chicken BEST1 orthologs highlighted conserved calcium-binding and gating mechanisms, with distinct features in mBEST1, including a wider aperture sufficient to accommodate dehydrated Cl- ions and potential anion-binding sites near Val205 and Gln208 residues. The disordered C-terminal region of mBEST1 remains unresolved, suggesting it may require stabilizing factors for structural determination. Additionally, the autoinhibitory domain, which includes Ser354, likely plays a key role in regulating gating, with Ser354 potentially serving as a phosphorylation site that modulates channel activity. Our findings provide structural and functional insights into mBEST1 and suggest mechanisms underlying its unique gating and ion permeation properties.
Collapse
Affiliation(s)
- Kwon-Woo Kim
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Euna Lee
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea; Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Ara Ko
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea; Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Junmo Hwang
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Kunwoong Park
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Byoung-Cheol Lee
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Ki Woo Kim
- Division of Physiology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul 03722, Republic of Korea
| | - Won-Jong Oh
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea
| | - Kyuhyung Kim
- Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Hyun-Ho Lim
- Neurovascular Unit Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Republic of Korea; Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea.
| |
Collapse
|
2
|
Ringlet S, Motta Z, Vandries L, Seutin V, Jehasse K, Caldinelli L, Pollegioni L, Engel D. Glycine-gated extrasynaptic NMDARs activated during glutamate spillover drive burst firing in nigral dopamine neurons. Prog Neurobiol 2025; 249:102773. [PMID: 40294743 DOI: 10.1016/j.pneurobio.2025.102773] [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: 12/19/2024] [Revised: 03/17/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Burst firing in substantia nigra pars compacta dopamine neurons is a critical biomarker temporally associated to movement initiation. This phasic change is generated by the tonic activation of NMDARs but the respective role of synaptic versus extrasynaptic NMDARs in the ignition of a burst and what is their level of activation remains unknown. Using ex vivo electrophysiological recordings from adolescent rats, we demonstrate that extrasynaptic NMDARs are the primary driver of burst firing. This pool of receptors is recruited during intense synaptic activity via spillover of glutamate and require the binding of NMDAR co-agonist glycine for full activation. Basal synaptic transmission activating only synaptic NMDARs with the support of D-serine is insufficient to generate a burst. Notably, both synaptic and extrasynaptic NMDARs share the same subunit composition but are regulated by distinct co-agonists. Location of NMDARs and regionalization of co-agonists but not NMDAR subunit composition underly burst generation and may serve as a guideline in understanding the physiological role of dopamine in signaling movement.
Collapse
Affiliation(s)
- Sofian Ringlet
- GIGA-Neurosciences, Laboratory of Molecular Regulation of Neurogenesis, University of Liege, Avenue Hippocrate 15, Liege B-4000, Belgium; GIGA-Neurosciences, Neurophysiology group, University of Liege, Avenue Hippocrate 15, Liege B-4000, Belgium
| | - Zoraide Motta
- The Protein Factory 2.0 Lab, Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, via Dunant, Varese 3-21100, Italy
| | - Laura Vandries
- GIGA-Neurosciences, Neurophysiology group, University of Liege, Avenue Hippocrate 15, Liege B-4000, Belgium
| | - Vincent Seutin
- GIGA-Neurosciences, Neurophysiology group, University of Liege, Avenue Hippocrate 15, Liege B-4000, Belgium
| | - Kevin Jehasse
- Montefiore Institute of Electrical Engineering and Computer Science, Systems and Modeling research unit at University of Liège, Quartier Polytech 1, allée de la Découverte 10, Liège 4000, Belgium
| | - Laura Caldinelli
- The Protein Factory 2.0 Lab, Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, via Dunant, Varese 3-21100, Italy
| | - Loredano Pollegioni
- The Protein Factory 2.0 Lab, Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell'Insubria, via Dunant, Varese 3-21100, Italy
| | - Dominique Engel
- GIGA-Neurosciences, Laboratory of Molecular Regulation of Neurogenesis, University of Liege, Avenue Hippocrate 15, Liege B-4000, Belgium.
| |
Collapse
|
3
|
Zhou B, Li Q, Su M, Liao P, Luo Y, Luo R, Yu Y, Luo M, Lei F, Li X, Jiao J, Yi L, Wang J, Yang L, Liao D, Zhou C, Zhang X, Xiao H, Zuo Y, Liu J, Zhu T, Jiang R. Astrocyte morphological remodeling regulates consciousness state transitions induced by inhaled general anesthesia. Mol Psychiatry 2025:10.1038/s41380-025-02978-2. [PMID: 40169801 DOI: 10.1038/s41380-025-02978-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025]
Abstract
General anesthetics (GAs) are conventionally thought to induce loss of consciousness (LOC) by acting on pre- and post-synaptic targets. However, the mechanism underlying the involvement of astrocytes in LOC remains unclear. Here we report that inhaled GAs cause reversible impairments in the fine processes of astrocytes within the somatosensory cortex, mediated by regulating the phosphorylation level of Ezrin, a protein critical for the fine morphology of astrocytes. Genetically deleting Ezrin or disrupting its phosphorylation was sufficient to decrease astrocyte-synapse interaction and enhance sensitivity to sevoflurane (Sevo) in vivo. Moreover, we show that disrupting astrocytic Ezrin phosphorylation boosted the inhibitory effect of Sevo on pyramidal neurons by enhancing tonic GABA and lowering excitability under anesthesia. Our work reveals previously unappreciated phosphorylation-dependent morphological dynamics, which enable astrocytes to regulate neuronal activity during the transition between two brain consciousness states.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qingran Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengchan Su
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Pain Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuncheng Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rong Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunqing Yu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Meiyan Luo
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fan Lei
- Institute of Brain Science and Diseases, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Xin Li
- Institute of Brain Science and Diseases, West China Hospital, Sichuan University, Chengdu, 610213, China
| | - Jiao Jiao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Limei Yi
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Wang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linghui Yang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Daqing Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cheng Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xia Zhang
- Institute of Brain Science and Diseases, West China Hospital, Sichuan University, Chengdu, 610213, China
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Xiao
- Department of Pain Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunxia Zuo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Song S, Kang M, Lee J, Yang YR, Lee H, Kim JI, Kim B, Choi HS, Hong EB, Nam MH, Suh PG, Kim J. Role of phospholipase Cη1 in lateral habenula astrocytes in depressive-like behavior in mice. Exp Mol Med 2025; 57:872-887. [PMID: 40204881 PMCID: PMC12046024 DOI: 10.1038/s12276-025-01432-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 04/11/2025] Open
Abstract
Phospholipase C (PLC) enzymes play crucial roles in intracellular calcium-signaling transduction. Several brain PLC subtypes have been extensively studied, implicating them in psychiatric disorders such as depression, epilepsy and schizophrenia. However, the role of the recently identified PLCη remains largely unknown. We found that PLCη1 is prominently expressed in lateral habenula (LHb) astrocytes. Here, to investigate its physiological role, we generated astrocyte-specific PLCη1 conditional knockout (cKO) mice (Plch1f/f; Aldh1l1-CreERT2). In these cKO mice, we observed a reduction in cellular morphological complexity metrics, such as total process length, as well as a decrease in the passive membrane conductance of LHb astrocytes. Additionally, neuronal function was impacted by the cKO, as the synaptic efficacy and firing rates of LHb neurons increased, while extrasynaptic long-term depression was impaired. Both tonic α-amino-3-hydroxy-5-methyl-4-isoxazolepdlropionic acid receptor/N-methyl-D-aspartate receptor (AMPAR/NMDAR) currents and extracellular glutamate levels were reduced. Interestingly, chemogenetic activation of astrocytes restored the reduced tonic AMPAR/NMDAR currents in cKO mice. Furthermore, LHb astrocyte-specific deletion of PLCη1 via AAV-GFAP-Cre injection induced depressive-like behaviors in mice, which were reversed by chemogenetic activation of LHb astrocytes. Finally, we found that restraint stress exposure decreased Plch1 mRNA expression in the LHb. These findings suggest that PLCη1 could be a potential therapeutic target for depression and highlight the critical role of astrocytes in the etiology of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sukwoon Song
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Miseon Kang
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jiyoung Lee
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Yong Ryoul Yang
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Ho Lee
- Cancer Experimental Resources Branch, National Cancer Center, Goyang, Republic of Korea
| | - Jae-Ick Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Beomsue Kim
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hoon-Seong Choi
- Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Eun-Bin Hong
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Pann-Ghill Suh
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jeongyeon Kim
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea.
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
5
|
Xiong S, Sun M, Zhang Y, Kong PR, Gan L, Gao L, Xu K, Wu HY, Zhu DY, Lin YH, Li R, Luo CX. Astrocytic BEST1 can serve as a target for functional recovery after ischemic stroke. Mol Ther 2025:S1525-0016(25)00196-0. [PMID: 40119514 DOI: 10.1016/j.ymthe.2025.03.022] [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: 08/03/2024] [Revised: 12/23/2024] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
Solid evidence from animal experiments supported the concept of peri-infarct tonic inhibition. Related drug targets have the potential to be translated for clinical stroke treatment. Recently, we reported the contribution of neuronal bestrophin-1 (BEST1)-mediated glutamate release to acute ischemic damage exacerbation in rodents. Now, we found a switch of abnormal BEST1 expression and function from neurons to astrocytes in the peri-infarct cortex following astrocytic activation. Excessive GABA was released through astrocytic BEST1 channel during the subacute phase of stroke, leading to sustained tonic inhibition. Astrocyte-specific knockdown of BEST1 promoted motor functional recovery, depending on reduced tonic inhibition. Moreover, we prepared self-assembled nanoparticles encapsulating siBest1 (SNP-siBest1), which displayed high brain accumulation and long circulation and knocked down astrocytic BEST1 effectively and safely. Systemic treatment with SNP-siBest1 after ischemic stroke showed a therapeutic effect in mice. Therefore, BEST1 is a potential target for stroke therapy from acute to subacute phase, and selective BEST1 blockers beyond nanoparticles are worth developing.
Collapse
Affiliation(s)
- Shuai Xiong
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Meng Sun
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ying Zhang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Pei-Ran Kong
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lu Gan
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ling Gao
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ke Xu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hai-Yin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Dong-Ya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yu-Hui Lin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Rui Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Chun-Xia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China; The Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
6
|
Faustmann TJ, Corvace F, Faustmann PM, Ismail FS. Influence of antipsychotic drugs on microglia-mediated neuroinflammation in schizophrenia: perspectives in an astrocyte-microglia co-culture model. Front Psychiatry 2025; 16:1522128. [PMID: 40171306 PMCID: PMC11959008 DOI: 10.3389/fpsyt.2025.1522128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/12/2025] [Indexed: 04/03/2025] Open
Abstract
Schizophrenia is a severe mental disorder with a strong lifetime impact on patients' health and wellbeing. Usually, symptomatic treatment includes typical or atypical antipsychotics. Study findings show an involvement of low-grade inflammation (blood, brain parenchyma, and cerebrospinal fluid) in schizophrenia. Moreover, experimental and neuropathological evidence suggests that reactive microglia, which are the main resident immune cells of the central nervous system (CNS), have a negative impact on the differentiation and function of oligodendrocytes, glial progenitor cells, and astrocytes, which results in the disruption of neuronal networks and dysregulated synaptic transmission, contributing to the pathophysiology of schizophrenia. Here, the role of microglial cells related to neuroinflammation in schizophrenia was discussed to be essential. This review aims to summarize the evidence for the influence of antipsychotics on microglial inflammatory mechanisms in schizophrenia. Furthermore, we propose an established astrocyte-microglia co-culture model for testing regulatory mechanisms and examining the effects of antipsychotics on glia-mediated neuroinflammation. This could lead to a better understanding of how typical and atypical antipsychotics can be used to address positive and negative symptoms in schizophrenia and comorbidities like inflammatory diseases or the status of low-grade inflammation.
Collapse
Affiliation(s)
- Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Franco Corvace
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Pedro M. Faustmann
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Fatme Seval Ismail
- Department of Neurology, Klinikum Vest, Academic Teaching Hospital of the Ruhr University Bochum, Recklinghausen, Germany
| |
Collapse
|
7
|
Kumari S, Bagri K, Deshmukh R. Connecting dots: Preclinical foundations to clinical realities of PDE4 inhibitors in Alzheimer's disease. Inflammopharmacology 2025; 33:593-603. [PMID: 39808238 DOI: 10.1007/s10787-024-01638-1] [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: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Alzheimer's Disease (AD), a progressive and age-associated neurodegenerative disorder, is primarily characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. Despite advances in targeting Aβ-mediated neuronal damage with anti-Aβ antibodies, these treatments provide only symptomatic relief and fail to address the multifactorial pathology of the disease. This necessitates the exploration of novel therapeutic approaches and a deeper understanding of molecular signaling mechanisms underlying AD. Phosphodiesterases (PDEs), particularly Phosphodiesterase 4 (PDE4), play a pivotal role in regulating cyclic adenosine monophosphate (cAMP), a key molecule involved in memory consolidation and cognitive function. PDE4 inhibitors have demonstrated potential in enhancing memory and cognition in preclinical models of AD by modulating cAMP signaling. However, their clinical translation has been limited due to challenges such as adverse effects, narrow therapeutic windows, and low specificity in mechanism of action. This review bridges the gap between preclinical discoveries and clinical applications of PDE4 inhibitors in AD. It highlights preclinical evidence supporting the neuroprotective and anti-inflammatory effects of PDE4 inhibitors while addressing challenges in their clinical development, including issues of safety, efficacy, and disease-specific targeting. By integrating findings from both preclinical and clinical studies, we provide a comprehensive understanding of the therapeutic potential of PDE4 inhibitors in AD. Furthermore, this review outlines future research directions aimed at optimizing PDE4 inhibition strategies for AD treatment, offering a roadmap to translate foundational insights into clinical realities.
Collapse
Affiliation(s)
- Shilpa Kumari
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, Punjab, India
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Kajal Bagri
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, Punjab, India
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Rahul Deshmukh
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India.
| |
Collapse
|
8
|
Juvenal G, Higa GSV, Bonfim Marques L, Tessari Zampieri T, Costa Viana FJ, Britto LR, Tang Y, Illes P, di Virgilio F, Ulrich H, de Pasquale R. Regulation of GABAergic neurotransmission by purinergic receptors in brain physiology and disease. Purinergic Signal 2025; 21:149-177. [PMID: 39046648 PMCID: PMC11958915 DOI: 10.1007/s11302-024-10034-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: 02/28/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Purinergic receptors regulate the processing of neural information in the hippocampus and cerebral cortex, structures related to cognitive functions. These receptors are activated when astrocytic and neuronal populations release adenosine triphosphate (ATP) in an autocrine and paracrine manner, following sustained patterns of neuronal activity. The modulation by these receptors of GABAergic transmission has only recently been studied. Through their ramifications, astrocytes and GABAergic interneurons reach large groups of excitatory pyramidal neurons. Their inhibitory effect establishes different synchronization patterns that determine gamma frequency rhythms, which characterize neural activities related to cognitive processes. During early life, GABAergic-mediated synchronization of excitatory signals directs the experience-driven maturation of cognitive development, and dysfunctions concerning this process have been associated with neurological and neuropsychiatric diseases. Purinergic receptors timely modulate GABAergic control over ongoing neural activity and deeply affect neural processing in the hippocampal and neocortical circuitry. Stimulation of A2 receptors increases GABA release from presynaptic terminals, leading to a considerable reduction in neuronal firing of pyramidal neurons. A1 receptors inhibit GABAergic activity but only act in the early postnatal period when GABA produces excitatory signals. P2X and P2Y receptors expressed in pyramidal neurons reduce the inhibitory tone by blocking GABAA receptors. Finally, P2Y receptor activation elicits depolarization of GABAergic neurons and increases GABA release, thus favoring the emergence of gamma oscillations. The present review provides an overall picture of purinergic influence on GABAergic transmission and its consequences on neural processing, extending the discussion to receptor subtypes and their involvement in the onset of brain disorders, including epilepsy and Alzheimer's disease.
Collapse
Affiliation(s)
- Guilherme Juvenal
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Guilherme Shigueto Vilar Higa
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lucas Bonfim Marques
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Thais Tessari Zampieri
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Felipe José Costa Viana
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luiz R Britto
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Yong Tang
- International Joint Research Centre On Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Peter Illes
- International Joint Research Centre On Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04107, Leipzig, Germany
| | | | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.
- International Joint Research Centre On Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Roberto de Pasquale
- Department of Biophysics and Physiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
9
|
Zinsmaier AK, Nestler EJ, Dong Y. Astrocytic G Protein-Coupled Receptors in Drug Addiction. ENGINEERING (BEIJING, CHINA) 2025; 44:256-265. [PMID: 40109668 PMCID: PMC11922559 DOI: 10.1016/j.eng.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Understanding the cellular mechanisms of drug addiction remains a key task in current brain research. While neuron-based mechanisms have been extensively explored over the past three decades, recent evidence indicates a critical involvement of astrocytes, the main type of non-neuronal cells in the brain. In response to extracellular stimuli, astrocytes modulate the activity of neurons, synaptic transmission, and neural network properties, collectively influencing brain function. G protein-coupled receptors (GPCRs) expressed on astrocyte surfaces respond to neuron- and environment-derived ligands by activating or inhibiting astrocytic signaling, which in turn regulates adjacent neurons and their circuitry. In this review, we focus on the dopamine D1 receptors (D1R) and metabotropic glutamate receptor 5 (mGLUR5 or GRM5)-two GPCRs that have been critically implicated in the acquisition and maintenance of addiction-related behaviors. Positioned as an introductory-level review, this article briefly discusses astrocyte biology, outlines earlier discoveries about the role of astrocytes in substance-use disorders (SUDs), and provides detailed discussion about astrocytic D1Rs and mGLUR5s in regulating synapse and network functions in the nucleus accumbens (NAc)-a brain region that mediates addiction-related emotional and motivational responses. This review serves as a stepping stone for readers of Engineering to explore links between astrocytic GPCRs and drug addiction and other psychiatric disorders.
Collapse
Affiliation(s)
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Yan Dong
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
10
|
Cheong E, Lee CJ. Gliotransmission in physiologic and pathologic conditions. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:93-116. [PMID: 40122634 DOI: 10.1016/b978-0-443-19104-6.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter explores the roles of gliotransmission in physiologic and pathologic conditions, including psychiatric and neurologic disorders. Gliotransmission, facilitated by astrocytes through the release of gliotransmitters such as glutamate, d-serine, and GABA, regulates neuronal activity and synaptic transmission. Under physiologic conditions, astrocytic gliotransmission maintains the balance of tonic excitation and inhibition, influencing synaptic plasticity and cognitive functions. In psychiatric disorders, the chapter examines how dysregulated gliotransmission contributes to major depression and schizophrenia. In major depression, changes in astrocytic glutamate and adenosine signaling impact mood regulation and cognitive functions. Schizophrenia involves complex astrocyte-neuron interactions, with dysregulated astrocytic activity affecting synaptic function and contributing to symptoms. The chapter also delves into neurologic disorders. In Alzheimer disease, aberrant GABA release from reactive astrocytes impairs memory and cognitive functions. Parkinson disease features alterations in glutamatergic and GABAergic systems, affecting motor and nonmotor symptoms. Epilepsy involves a disruption in the balance between excitatory and inhibitory neurotransmission, with astrocytic GABA accumulation helping to maintain neuronal stability. Autism spectrum disorder (ASD) is linked to imbalances in glutamatergic and GABAergic neurotransmission, underlying sensory, cognitive, and social impairments. Overall, the chapter underscores the pivotal role of gliotransmission in maintaining neural homeostasis and highlights its potential as a therapeutic target in various disorders.
Collapse
Affiliation(s)
- Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea.
| |
Collapse
|
11
|
Pál B. On the functions of astrocyte-mediated neuronal slow inward currents. Neural Regen Res 2024; 19:2602-2612. [PMID: 38595279 PMCID: PMC11168512 DOI: 10.4103/nrr.nrr-d-23-01723] [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: 10/17/2023] [Revised: 12/25/2023] [Accepted: 01/24/2024] [Indexed: 04/11/2024] Open
Abstract
Slow inward currents are known as neuronal excitatory currents mediated by glutamate release and activation of neuronal extrasynaptic N-methyl-D-aspartate receptors with the contribution of astrocytes. These events are significantly slower than the excitatory postsynaptic currents. Parameters of slow inward currents are determined by several factors including the mechanisms of astrocytic activation and glutamate release, as well as the diffusion pathways from the release site towards the extrasynaptic receptors. Astrocytes are stimulated by neuronal network activity, which in turn excite neurons, forming an astrocyte-neuron feedback loop. Mostly as a consequence of brain edema, astrocytic swelling can also induce slow inward currents under pathological conditions. There is a growing body of evidence on the roles of slow inward currents on a single neuron or local network level. These events often occur in synchrony on neurons located in the same astrocytic domain. Besides synchronization of neuronal excitability, slow inward currents also set synaptic strength via eliciting timing-dependent synaptic plasticity. In addition, slow inward currents are also subject to non-synaptic plasticity triggered by long-lasting stimulation of the excitatory inputs. Of note, there might be important region-specific differences in the roles and actions triggering slow inward currents. In greater networks, the pathophysiological roles of slow inward currents can be better understood than physiological ones. Slow inward currents are identified in the pathophysiological background of autism, as slow inward currents drive early hypersynchrony of the neural networks. Slow inward currents are significant contributors to paroxysmal depolarizational shifts/interictal spikes. These events are related to epilepsy, but also found in Alzheimer's disease, Parkinson's disease, and stroke, leading to the decline of cognitive functions. Events with features overlapping with slow inward currents (excitatory, N-methyl-D-aspartate-receptor mediated currents with astrocytic contribution) as ischemic currents and spreading depolarization also have a well-known pathophysiological role in worsening consequences of stroke, traumatic brain injury, or epilepsy. One might assume that slow inward currents occurring with low frequency under physiological conditions might contribute to synaptic plasticity and memory formation. However, to state this, more experimental evidence from greater neuronal networks or the level of the individual is needed. In this review, I aimed to summarize findings on slow inward currents and to speculate on the potential functions of it.
Collapse
Affiliation(s)
- Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
12
|
Andrade-Talavera Y, Sánchez-Gómez J, Coatl-Cuaya H, Rodríguez-Moreno A. Developmental Spike Timing-Dependent Long-Term Depression Requires Astrocyte d-Serine at L2/3-L2/3 Synapses of the Mouse Somatosensory Cortex. J Neurosci 2024; 44:e0805242024. [PMID: 39406518 PMCID: PMC11604139 DOI: 10.1523/jneurosci.0805-24.2024] [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: 04/30/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 11/29/2024] Open
Abstract
Spike timing-dependent plasticity (STDP) is a learning rule important for synaptic refinement and for learning and memory during development. While different forms of presynaptic t-LTD have been deeply investigated, little is known about the mechanisms of somatosensory cortex postsynaptic t-LTD. In the present work, we investigated the requirements and mechanisms for induction of developmental spike timing-dependent long-term depression (t-LTD) at L2/3-L2/3 synapses in the juvenile mouse somatosensory cortex. We found that postnatal day (P) 13-21 mice of either sex show t-LTD at L2/3-L2/3 synapses induced by pairing single presynaptic activity with single postsynaptic action potentials at low stimulation frequency (0.2 Hz) that is expressed postsynaptically and requires the activation of ionotropic postsynaptic NMDA-type glutamate receptors containing GluN2B subunits. In addition, it requires postsynaptic Ca2+, Ca2+ release from internal stores, calcineurin, postsynaptic endocannabinoid synthesis, activation of CB1 receptors, and astrocytic signaling to release the gliotransmitter d-serine to activate postsynaptic NMDARs to induce t-LTD. These results show direct evidence of the mechanism involved in developmental postsynaptic t-LTD at L2/3-L2/3 synapses, revealing a central role of astrocytes and their release of d-serine in its induction.
Collapse
Affiliation(s)
- Yuniesky Andrade-Talavera
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Seville ES-41013, Spain
| | - Joaquín Sánchez-Gómez
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Seville ES-41013, Spain
| | - Heriberto Coatl-Cuaya
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Seville ES-41013, Spain
| | - Antonio Rodríguez-Moreno
- Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, Seville ES-41013, Spain
| |
Collapse
|
13
|
Parodi J, Mira RG, Fuenzalida M, Cerpa W, Serrano FG, Tapia-Rojas C, Martinez-Torres A, Inestrosa NC. Wnt-5a Signaling Mediates Metaplasticity at Hippocampal CA3-CA1 Synapses in Mice. Cell Mol Neurobiol 2024; 44:76. [PMID: 39535658 PMCID: PMC11561030 DOI: 10.1007/s10571-024-01512-2] [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: 08/22/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Wnt signaling plays a role in synaptic plasticity, but the specific cellular events and molecular components involved in Wnt signaling-mediated synaptic plasticity are not well defined. Here, we report a change in the threshold required to induce synaptic plasticity that facilitates the induction of long-term potentiation (LTP) and inhibits the induction of long-term depression (LTD) during brief exposure to the noncanonical ligand Wnt-5a. Both effects are related to the metaplastic switch of hippocampal CA3-CA1 synaptic transmission, a complex mechanism underlying the regulation of the threshold required to induce synaptic plasticity and of synaptic efficacy. We observed an early increase in the amplitude of field excitatory postsynaptic potentials (fEPSPs) that persisted over time, including after washout. The first phase involves an increase in the fEPSP amplitude that is required to trigger a spontaneous second phase that depends on Jun N-terminal kinase (JNK) and N-methyl D-aspartate receptor (NMDAR) activity. These changes are prevented by treatment with secreted frizzled-related protein 2 (sFRP-2), an endogenous antagonist of Wnt ligands. Here, we demonstrate the contribution of Wnt-5a signaling to a process associated with metaplasticity at CA3-CA1 synapses that favors LTP over LTD.
Collapse
Affiliation(s)
- Jorge Parodi
- Departamento de Análisis de Datos, Facultad de Ciencias Sociales, Universidad Autónoma de Chile, Temuco, Chile
| | - Rodrigo G Mira
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Millenium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Waldo Cerpa
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe G Serrano
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cheril Tapia-Rojas
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Ataulfo Martinez-Torres
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Nibaldo C Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Escuela de Medicina, Universidad de Magallanes, Punta Arenas, Chile.
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
14
|
Ma H, He S, Li Y, Zhang X, Chang H, Du M, Yan C, Jiang S, Gao H, Zhao J, Wang Q. Augmented Mitochondrial Transfer Involved in Astrocytic PSPH Attenuates Cognitive Dysfunction in db/db Mice. Mol Neurobiol 2024; 61:8872-8885. [PMID: 38573412 DOI: 10.1007/s12035-024-04064-0] [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: 07/31/2023] [Accepted: 02/19/2024] [Indexed: 04/05/2024]
Abstract
Diabetes-associated cognitive dysfunction (DACD) has ascended to become the second leading cause of mortality among diabetic patients. Phosphoserine phosphatase (PSPH), a pivotal rate-limiting enzyme in L-serine biosynthesis, has been documented to instigate the insulin signaling pathway through dephosphorylation. Concomitantly, CD38, acting as a mediator in mitochondrial transfer, is activated by the insulin pathway. Given that we have demonstrated the beneficial effects of exogenous mitochondrial supplementation on DACD, we further hypothesized whether astrocytic PSPH could contribute to improving DACD by promoting astrocytic mitochondrial transfer into neurons. In the Morris Water Maze (MWM) test, our results demonstrated that overexpression of PSPH in astrocytes alleviated DACD in db/db mice. Astrocyte specific-stimulated by PSPH lentivirus/ adenovirus promoted the spine density both in vivo and in vitro. Mechanistically, astrocytic PSPH amplified the expression of CD38 via initiation of the insulin signaling pathway, thereby promoting astrocytic mitochondria transfer into neurons. In summation, this comprehensive study delineated the pivotal role of astrocytic PSPH in alleviating DACD and expounded upon its intricate cellular mechanism involving mitochondrial transfer. These findings propose that the specific up-regulation of astrocytic PSPH holds promise as a discerning therapeutic modality for DACD.
Collapse
Affiliation(s)
- Hongli Ma
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Department of Anesthesiology, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100029, China
| | - Shuxuan He
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xin Zhang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Haiqing Chang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Mengyu Du
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Chaoying Yan
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Shiqiu Jiang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Hui Gao
- Department of Anesthesiology, Yan'an University Affiliated Hospital, Yan'an, Shaanxi, 716000, China
| | - Jing Zhao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Department of Anesthesiology, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100029, China.
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Department of Anesthesiology, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100029, China.
| |
Collapse
|
15
|
Wei N, Guo Z, Qiu M, Ye R, Shao X, Liang Y, Liu B, Fang J, Fang J, Du J. Astrocyte Activation in the ACC Contributes to Comorbid Anxiety in Chronic Inflammatory Pain and Involves in The Excitation-Inhibition Imbalance. Mol Neurobiol 2024; 61:6934-6949. [PMID: 38363535 DOI: 10.1007/s12035-024-04027-5] [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: 11/06/2023] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
Neurons within the anterior cingulate cortex (ACC) orchestrate the co-occurrence of chronic pain and anxiety. The ACC hyperactivity plays a crucial role in the emotional impact of neuropathic pain. Astrocyte-mediated neuroinflammatory is responsible for regulating the balance between excitation-inhibition (E/I) in the brain. However, there is limited understanding of the possible contributions of astrocytes in the ACC to comorbidity of anxiety and chronic inflammatory pain. This paper aims to investigate the possible contribution of astrocytes in the ACC to the comorbidity between anxiety and chronic inflammatory pain, as well as their involvement in the E/I imbalance of pyramidal cells. Our results show that CFA rats displayed allodynia and anxiety-like behaviors. The E/I balance in the ACC shifts to excitement in comorbidity of chronic pain and anxiety by western blotting, and electrophysiological recording. Result of RNA-Seq also indicated that E/I imbalance and neuroinflammation of ACC were involved in pain-anxiety comorbidity. Then, positive cells of GFAP but not Iba1 in the contralateral ACC were increased; the mRNA expression of GFAP and its activation-related proinflammatory cytokines (TNF-α, IL-6, and IL-1β) in the contralateral ACC were also elevated. Furthermore, specific chemogenic inhibition of ACC astrocytes reversed comorbid pain and anxiety and suppressed high ACC excitability. Our data suggest that astrocytes participate in comorbid pain and anxiety and excitation-inhibition imbalance in ACC. Inhibition astrocyte activation can reduce anxiety related to pain and restore the imbalance in the ACC. These findings shed light on the involvement of astrocytes in comorbid conditions, offering valuable insights into a potential therapeutic approach for the co-occurrence of chronic pain and anxiety.
Collapse
Affiliation(s)
- Naixuan Wei
- Department of Neurobiology and Acupuncture Research, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zi Guo
- Department of Neurobiology and Acupuncture Research, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengting Qiu
- Fuchun Community Health Service Center of Fuyang District, Hangzhou, 311400, China
| | - Ru Ye
- Department of Neurobiology and Acupuncture Research, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaomei Shao
- Department of Neurobiology and Acupuncture Research, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yi Liang
- Department of Neurobiology and Acupuncture Research, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Boyi Liu
- Department of Neurobiology and Acupuncture Research, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jianqiao Fang
- Department of Neurobiology and Acupuncture Research, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Junfan Fang
- Department of Neurobiology and Acupuncture Research, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Junying Du
- Department of Neurobiology and Acupuncture Research, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
16
|
Joo J, Kim KJ, Lim J, Choi SY, Koh W, Lee CJ. Generation of Astrocyte-specific BEST1 Conditional Knockout Mouse with Reduced Tonic GABA Inhibition in the Brain. Exp Neurobiol 2024; 33:180-192. [PMID: 39266474 PMCID: PMC11411089 DOI: 10.5607/en24019] [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: 08/15/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/14/2024] Open
Abstract
Bestrophin-1 (BEST1) is a Ca2+-activated anion channel known for its role in astrocytes. Best1 is permeable to gliotransmitters, including GABA, to contribute to tonic GABA inhibition and modulate synaptic transmission in neighboring neurons. Despite the crucial functions of astrocytic BEST1, there is an absence of genetically engineered cell-type specific conditional mouse models addressing these roles. In this study, we developed an astrocyte-specific BEST1 conditional knock-out (BEST1 aKO) mouse line. Using the embryonic stem cell (ES cell) targeting method, we developed Best1 floxed mice (C57BL/6JCya-Best1em1flox/Cya), which have exon 3, 4, 5, and 6 of Best1 flanked by two loxP sites. By crossing with hGFAP-CreERT2 mice, we generated Best1 floxed/hGFAP-CreERT2 mice, which allowed for the tamoxifen-inducible deletion of Best1 under the human GFAP promoter. We characterized its features across various brain regions, including the striatum, hippocampal dentate gyrus (HpDG), and Parafascicular thalamic nucleus (Pf). Compared to the Cre-negative control, we observed significantly reduced BEST1 protein expression in immunohistochemistry (IHC) and tonic GABA inhibition in patch clamp recordings. The reduction in tonic GABA inhibition was 66.7% in the striatum, 46.4% in the HpDG, and 49.6% in the Pf. Our findings demonstrate that the BEST1 channel in astrocytes significantly contributes to tonic inhibition in the local brain areas. These mice will be valuable for future studies not only on tonic GABA release but also on tonic release of gliotransmitters mediated by astrocytic BEST1.
Collapse
Affiliation(s)
- Jinhyeong Joo
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
- IBS School, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Ki Jung Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
- IBS School, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Sun Yeong Choi
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
- IBS School, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| |
Collapse
|
17
|
Kim H, Choi S, Lee E, Koh W, Lee CJ. Tonic NMDA Receptor Currents in the Brain: Regulation and Cognitive Functions. Biol Psychiatry 2024; 96:164-175. [PMID: 38490367 DOI: 10.1016/j.biopsych.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
Synaptically localized NMDA receptors (NMDARs) play a crucial role in important cognitive functions by mediating synaptic transmission and plasticity. In contrast, a tonic NMDAR current, thought to be mediated by extrasynaptic NMDARs, has a less clear function. This review provides a comprehensive overview of tonic NMDAR currents, focusing on their roles in synaptic transmission/plasticity and their impact on cognitive functions and psychiatric disorders. We discuss the roles of 3 endogenous ligands (i.e., glutamate, glycine, and D-serine) and receptors in mediating tonic NMDAR currents and explore the diverse mechanisms that regulate tonic NMDAR currents. In light of recent controversies surrounding the source of D-serine, we highlight the recent findings suggesting that astrocytes release D-serine to modulate tonic NMDAR currents and control cognitive flexibility. Furthermore, we propose distinct roles of neuronal and astrocytic D-serine in different locations and their implications for synaptic regulation and cognitive functions. The potential roles of tonic NMDAR currents in various psychiatric disorders, such as schizophrenia and autism spectrum disorder, are discussed in the context of the NMDAR hypofunction hypothesis. By presenting the mechanisms by which various cells, particularly astrocytes, regulate tonic NMDAR currents, we aim to stimulate future research in NMDAR hypofunction- or hyperfunction-related psychiatric disorders. This review not only provides a better understanding of the complex interplay between tonic NMDAR currents and cognitive functions but also sheds light on its potential therapeutic target for the treatment of various psychiatric disorders.
Collapse
Affiliation(s)
- Hayoung Kim
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science, Daejeon, South Korea
| | - Sunyeong Choi
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science, Daejeon, South Korea
| | - Euisun Lee
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science, Daejeon, South Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science, Daejeon, South Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science, Daejeon, South Korea.
| |
Collapse
|
18
|
Squadrani L, Wert-Carvajal C, Müller-Komorowska D, Bohmbach K, Henneberger C, Verzelli P, Tchumatchenko T. Astrocytes enhance plasticity response during reversal learning. Commun Biol 2024; 7:852. [PMID: 38997325 PMCID: PMC11245475 DOI: 10.1038/s42003-024-06540-8] [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: 08/26/2023] [Accepted: 07/03/2024] [Indexed: 07/14/2024] Open
Abstract
Astrocytes play a key role in the regulation of synaptic strength and are thought to orchestrate synaptic plasticity and memory. Yet, how specifically astrocytes and their neuroactive transmitters control learning and memory is currently an open question. Recent experiments have uncovered an astrocyte-mediated feedback loop in CA1 pyramidal neurons which is started by the release of endocannabinoids by active neurons and closed by astrocytic regulation of the D-serine levels at the dendrites. D-serine is a co-agonist for the NMDA receptor regulating the strength and direction of synaptic plasticity. Activity-dependent D-serine release mediated by astrocytes is therefore a candidate for mediating between long-term synaptic depression (LTD) and potentiation (LTP) during learning. Here, we show that the mathematical description of this mechanism leads to a biophysical model of synaptic plasticity consistent with the phenomenological model known as the BCM model. The resulting mathematical framework can explain the learning deficit observed in mice upon disruption of the D-serine regulatory mechanism. It shows that D-serine enhances plasticity during reversal learning, ensuring fast responses to changes in the external environment. The model provides new testable predictions about the learning process, driving our understanding of the functional role of neuron-glia interaction in learning.
Collapse
Affiliation(s)
- Lorenzo Squadrani
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Carlos Wert-Carvajal
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Kirsten Bohmbach
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Pietro Verzelli
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany.
| | - Tatjana Tchumatchenko
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
19
|
Lalo U, Pankratov Y. Astrocyte ryanodine receptors facilitate gliotransmission and astroglial modulation of synaptic plasticity. Front Cell Neurosci 2024; 18:1382010. [PMID: 38812795 PMCID: PMC11135129 DOI: 10.3389/fncel.2024.1382010] [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: 02/04/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Intracellular Ca2+-signaling in astrocytes is instrumental for their brain "housekeeping" role and astroglial control of synaptic plasticity. An important source for elevating the cytosolic Ca2+ level in astrocytes is a release from endoplasmic reticulum which can be triggered via two fundamental pathways: IP3 receptors and calcium-induced calcium release (CICR) mediated by Ca2+-sensitive ryanodine receptors (RyRs). While the physiological role for glial IP3 became a focus of intensive research and debate, ryanodine receptors received much less attention. We explored the role for ryanodine receptors in the modulation of cytosolic Ca2+-signaling in the cortical and hippocampal astrocytes, astrocyte-neuron communication and astroglia modulation of synaptic plasticity. Our data show that RyR-mediated Ca2+-induced Ca2+-release from ER brings substantial contribution into signaling in the functional microdomains hippocampal and neocortical astrocytes. Furthermore, RyR-mediated CICR activated the release of ATP and glutamate from hippocampal and neocortical astrocytes which, in turn, elicited transient purinergic and tonic glutamatergic currents in the neighboring pyramidal neurons. The CICR-facilitated release of ATP and glutamate was inhibited after intracellular perfusion of astrocytes with ryanodine and BAPTA and in the transgenic dnSNARE mice with impaired astroglial exocytosis. We also found out that RyR-mediated amplification of astrocytic Ca2+-signaling enhanced the long-term synaptic potentiation in the hippocampus and neocortex of aged mice. Combined, our data demonstrate that ryanodine receptors are essential for astrocytic Ca2+-signaling and efficient astrocyte-neuron communications. The RyR-mediated CICR contributes to astrocytic control of synaptic plasticity and can underlie, at least partially, neuroprotective and cognitive effects of caffein.
Collapse
Affiliation(s)
| | - Yuriy Pankratov
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
20
|
Kim HJ, Phan TT, Lee K, Kim JS, Lee SY, Lee JM, Do J, Lee D, Kim SP, Lee KP, Park J, Lee CJ, Park JM. Long-lasting forms of plasticity through patterned ultrasound-induced brainwave entrainment. SCIENCE ADVANCES 2024; 10:eadk3198. [PMID: 38394205 PMCID: PMC10889366 DOI: 10.1126/sciadv.adk3198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024]
Abstract
Achieving long-lasting neuronal modulation with low-intensity, low-frequency ultrasound is challenging. Here, we devised theta burst ultrasound stimulation (TBUS) with gamma bursts for brain entrainment and modulation of neuronal plasticity in the mouse motor cortex. We demonstrate that two types of TBUS, intermittent and continuous TBUS, induce bidirectional long-term potentiation or depression-like plasticity, respectively, as evidenced by changes in motor-evoked potentials. These effects depended on molecular pathways associated with long-term plasticity, including N-methyl-d-aspartate receptor and brain-derived neurotrophic factor/tropomyosin receptor kinase B activation, as well as de novo protein synthesis. Notably, bestrophin-1 and transient receptor potential ankyrin 1 play important roles in these enduring effects. Moreover, pretraining TBUS enhances the acquisition of previously unidentified motor skills. Our study unveils a promising protocol for ultrasound neuromodulation, enabling noninvasive and sustained modulation of brain function.
Collapse
Affiliation(s)
- Ho-Jeong Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Tien Thuy Phan
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Keunhyung Lee
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jeong Sook Kim
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sang-Yeong Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jung Moo Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jongrok Do
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Doyun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Sung-Phil Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Kyu Pil Lee
- Department of Physiology, College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jinhyoung Park
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - C. Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
- University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Joo Min Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
- University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
21
|
Imrie G, Gray MB, Raghuraman V, Farhy-Tselnicker I. Gene Expression at the Tripartite Synapse: Bridging the Gap Between Neurons and Astrocytes. ADVANCES IN NEUROBIOLOGY 2024; 39:95-136. [PMID: 39190073 DOI: 10.1007/978-3-031-64839-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Astrocytes, a major class of glial cells, are an important element at the synapse where they engage in bidirectional crosstalk with neurons to regulate numerous aspects of neurotransmission, circuit function, and behavior. Mutations in synapse-related genes expressed in both neurons and astrocytes are central factors in a vast number of neurological disorders, making the proteins that they encode prominent targets for therapeutic intervention. Yet, while the roles of many of these synaptic proteins in neurons are well established, the functions of the same proteins in astrocytes are largely unknown. This gap in knowledge must be addressed to refine therapeutic approaches. In this chapter, we integrate multiomic meta-analysis and a comprehensive overview of current literature to show that astrocytes express an astounding number of genes that overlap with the neuronal and synaptic transcriptomes. Further, we highlight recent reports that characterize the expression patterns and potential novel roles of these genes in astrocytes in both physiological and pathological conditions, underscoring the importance of considering both cell types when investigating the function and regulation of synaptic proteins.
Collapse
Affiliation(s)
- Gillian Imrie
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Madison B Gray
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Vishnuvasan Raghuraman
- Department of Biology, Texas A&M University, College Station, TX, USA
- Department of Computer Science and Engineering, Texas A&M University, College Station, TX, USA
| | - Isabella Farhy-Tselnicker
- Department of Biology, Texas A&M University, College Station, TX, USA.
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, USA.
- Center for Biological Clocks Research, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
22
|
Abreu DS, Gomes JI, Ribeiro FF, Diógenes MJ, Sebastião AM, Vaz SH. Astrocytes control hippocampal synaptic plasticity through the vesicular-dependent release of D-serine. Front Cell Neurosci 2023; 17:1282841. [PMID: 38145284 PMCID: PMC10740624 DOI: 10.3389/fncel.2023.1282841] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/10/2023] [Indexed: 12/26/2023] Open
Abstract
Astrocytes, the most abundant glial cells in the central nervous system (CNS), sense synaptic activity and respond through the release of gliotransmitters, a process mediated by intracellular Ca2+ level changes and SNARE-dependent mechanisms. Ionotropic N-methyl-D-aspartate (NMDA) receptors, which are activated by glutamate along with D-serine or glycine, play a crucial role in learning, memory, and synaptic plasticity. However, the precise impact of astrocyte-released D-serine on neuronal modulation remains insufficiently characterized. To address this, we have used the dominant negative SNARE (dnSNARE) mouse model, which selectively inhibits SNARE-dependent exocytosis from astrocytes. We recorded field excitatory postsynaptic potentials (fEPSPs) in CA3-CA1 synapses within hippocampal slices obtained from dnSNARE mice and wild-type (Wt) littermates. Our results demonstrate that hippocampal θ-burst long-term potentiation (LTP), a critical form of synaptic plasticity, is impaired in hippocampal slices from dnSNARE mice. Notably, this LTP impairment was rescued upon incubation with D-serine. To further investigate the involvement of astrocytes in D-serine-mediated mechanisms of LTP maintenance, we perfused hippocampal slices with L-serine - a substrate used by both neurons and astrocytes for D-serine production. The enhancement in LTP observed in dnSNARE mice was exclusively associated with D-serine presence, with no effects evident in the presence of L-serine. Additionally, both D- and L-serine reduced basal synaptic strength in the hippocampal slices of both Wt and dnSNARE mice. These results provide compelling evidence that distinct processes underlie the modulation of basal synaptic transmission and LTP through D-serine. Our findings underscore the pivotal contribution of astrocytes in D-serine-mediated processes that govern LTP establishment and basal transmission. This study not only provides essential insights into the intricate interplay between neurons and astrocytes but also emphasizes their collective role in shaping hippocampal synaptic function.
Collapse
Affiliation(s)
- Daniela Sofia Abreu
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisbon, Portugal
| | - Joana I. Gomes
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisbon, Portugal
| | - Filipa F. Ribeiro
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisbon, Portugal
| | - Maria J. Diógenes
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisbon, Portugal
| | - Ana M. Sebastião
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisbon, Portugal
| | - Sandra H. Vaz
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
23
|
Mony L, Paoletti P. Mechanisms of NMDA receptor regulation. Curr Opin Neurobiol 2023; 83:102815. [PMID: 37988826 DOI: 10.1016/j.conb.2023.102815] [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: 09/13/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/23/2023]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are glutamate-gated ion channels widely expressed in the central nervous system that play key role in brain development and plasticity. On the downside, NMDAR dysfunction, be it hyperactivity or hypofunction, is harmful to neuronal function and has emerged as a common theme in various neuropsychiatric disorders including autism spectrum disorders, epilepsy, intellectual disability, and schizophrenia. Not surprisingly, NMDAR signaling is under a complex set of regulatory mechanisms that maintain NMDAR-mediated transmission in check. These include an unusual large number of endogenous agents that directly bind NMDARs and tune their activity in a subunit-dependent manner. Here, we review current knowledge on the regulation of NMDAR signaling. We focus on the regulation of the receptor by its microenvironment as well as by external (i.e. pharmacological) factors and their underlying molecular and cellular mechanisms. Recent developments showing how NMDAR dysregulation participate to disease mechanisms are also highlighted.
Collapse
Affiliation(s)
- Laetitia Mony
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005 Paris, France.
| | - Pierre Paoletti
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005 Paris, France.
| |
Collapse
|
24
|
Cha H, Choi JH, Jeon H, Kim JH, Kim M, Kim SJ, Park W, Lim JS, Lee E, Ahn JS, Kim JH, Hong SH, Park JE, Jung JH, Yoo HJ, Lee S. Aquaporin-4 Deficiency is Associated with Cognitive Impairment and Alterations in astrocyte-neuron Lactate Shuttle. Mol Neurobiol 2023; 60:6212-6226. [PMID: 37436602 DOI: 10.1007/s12035-023-03475-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/02/2023] [Indexed: 07/13/2023]
Abstract
Cognitive impairment refers to notable declines in cognitive abilities including memory, language, and emotional stability leading to the inability to accomplish essential activities of daily living. Astrocytes play an important role in cognitive function, and homeostasis of the astrocyte-neuron lactate shuttle (ANLS) system is essential for maintaining cognitive functions. Aquaporin-4 (AQP-4) is a water channel expressed in astrocytes and has been shown to be associated with various brain disorders, but the direct relationship between learning, memory, and AQP-4 is unclear. We examined the relationship between AQP-4 and cognitive functions related to learning and memory. Mice with genetic deletion of AQP-4 showed significant behavioral and emotional changes including hyperactivity and instability, and impaired cognitive functions such as spatial learning and memory retention. 18 F-FDG PET imaging showed significant metabolic changes in the brains of AQP-4 knockout mice such as reductions in glucose absorption. Such metabolic changes in the brain seemed to be the direct results of changes in the expression of metabolite transporters, as the mRNA levels of multiple glucose and lactate transporters in astrocytes and neurons were significantly decreased in the cortex and hippocampus of AQP-4 knockout mice. Indeed, AQP-4 knockout mice showed significantly higher accumulation of both glucose and lactate in their brains compared with wild-type mice. Our results show that the deficiency of AQP-4 can cause problems in the metabolic function of astrocytes and lead to cognitive impairment, and that the deficiency of AQP4 in astrocyte endfeet can cause abnormalities in the ANLS system.
Collapse
Affiliation(s)
- Hyeuk Cha
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jun Ho Choi
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hanwool Jeon
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae Hyun Kim
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Moinay Kim
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Su Jung Kim
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Wonhyoung Park
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joon Seo Lim
- Clinical Research Center, Asan Medical Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eunyeup Lee
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae Sung Ahn
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong Hoon Kim
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seok Ho Hong
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
- University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Park
- University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Neuroradiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Hwa Jung
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Hyun Ju Yoo
- University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Seungjoo Lee
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Department of Medical Science, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Li N, Ren C, Li S, Yu W, Jin K, Ji X. Remote ischemic conditioning alleviates chronic cerebral hypoperfusion-induced cognitive decline and synaptic dysfunction via the miR-218a-5p/SHANK2 pathway. Prog Neurobiol 2023; 230:102514. [PMID: 37574039 DOI: 10.1016/j.pneurobio.2023.102514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/15/2023]
Abstract
Vascular cognitive impairment (VCI) due to chronic cerebral hypoperfusion (CCH), is the second leading cause of dementia. Although synaptic impairment plays a critical role in VCI, its exact mechanism remains unknown. Our previous research revealed that remote ischemic conditioning (RIC) could alleviate cognitive decline resulting from CCH, however, its effects on synaptic impairment remain unclear. In this study, we confirmed that RIC alleviated both cognitive decline and its associated synaptic dysfunction caused by CCH. RNA sequencing revealed that CCH increased in miR-218a-5p expression, which was decreased by RIC. Elevated miR-218a-5p levels limited the benefits of RIC, however, inhibiting miR-218a-5p in hippocampal CA1 neurons rescued synaptic dysfunction. Additionally, we found that SHANK2 is a downstream target of miR-218a-5p, and inhibiting SHANK2 expression reduced the alleviation caused by hypoxic conditioning in synaptic impairment in vitro. In conclusion, our results suggested that RIC alleviated synaptic impairment via the miR-218a-5p/SHANK2 pathway, which could be a potential biomarker or therapeutic target for cognitive impairment caused by CCH.
Collapse
Affiliation(s)
- Ning Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorder, Capital Medical University, Beijing 100053, China
| | - Wantong Yu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Kunlin Jin
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Xuming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorder, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
26
|
Zhang F, Liu M, Tuo J, Zhang L, Zhang J, Yu C, Xu Z. Levodopa-induced dyskinesia: interplay between the N-methyl-D-aspartic acid receptor and neuroinflammation. Front Immunol 2023; 14:1253273. [PMID: 37860013 PMCID: PMC10582719 DOI: 10.3389/fimmu.2023.1253273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder of middle-aged and elderly people, clinically characterized by resting tremor, myotonia, reduced movement, and impaired postural balance. Clinically, patients with PD are often administered levodopa (L-DOPA) to improve their symptoms. However, after years of L-DOPA treatment, most patients experience complications of varying severity, including the "on-off phenomenon", decreased efficacy, and levodopa-induced dyskinesia (LID). The development of LID can seriously affect the quality of life of patients, but its pathogenesis is unclear and effective treatments are lacking. Glutamic acid (Glu)-mediated changes in synaptic plasticity play a major role in LID. The N-methyl-D-aspartic acid receptor (NMDAR), an ionotropic glutamate receptor, is closely associated with synaptic plasticity, and neuroinflammation can modulate NMDAR activation or expression; in addition, neuroinflammation may be involved in the development of LID. However, it is not clear whether NMDA receptors are co-regulated with neuroinflammation during LID formation. Here we review how neuroinflammation mediates the development of LID through the regulation of NMDA receptors, and assess whether common anti-inflammatory drugs and NMDA receptor antagonists may be able to mitigate the development of LID through the regulation of central neuroinflammation, thereby providing a new theoretical basis for finding new therapeutic targets for LID.
Collapse
Affiliation(s)
- Fanshi Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Mei Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jinmei Tuo
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Li Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Changyin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| |
Collapse
|
27
|
Tripodi F, Motta Z, Murtas G, Rabattoni V, Nonnis S, Grassi Scalvini F, Rinaldi AM, Rizzi R, Bearzi C, Badone B, Sacchi S, Tedeschi G, Maffioli E, Coccetti P, Pollegioni L. Serine metabolism during differentiation of human iPSC-derived astrocytes. FEBS J 2023; 290:4440-4464. [PMID: 37166453 DOI: 10.1111/febs.16816] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/05/2023] [Accepted: 05/10/2023] [Indexed: 05/12/2023]
Abstract
Astrocytes are essential players in development and functions, being particularly relevant as regulators of brain energy metabolism, ionic homeostasis and synaptic transmission. They are also the major source of l-serine in the brain, which is synthesized from the glycolytic intermediate 3-phosphoglycerate through the phosphorylated pathway. l-Serine is the precursor of the two main co-agonists of the N-methyl-d-aspartate receptor, glycine and d-serine. Strikingly, dysfunctions in both l- and d-serine metabolism are associated with neurological and psychiatric disorders. Here, we exploited a differentiation protocol, based on the generation of human mature astrocytes from neural stem cells, and investigated the modification of the proteomic and metabolomic profile during the differentiation process. We show that differentiated astrocytes are more similar to mature rather than to reactive ones, and that axogenesis and pyrimidine metabolism increase up to 30 days along with the folate cycle and sphingolipid metabolism. Consistent with the proliferation and cellular maturation processes that are taking place, also the intracellular levels of l-serine, glycine, threonine, l- and d-aspartate (which level is unexpectedly higher than that of d-serine) show the same biosynthetic time course. A significant utilization of l-serine from the medium is apparent while glycine is first consumed and then released with a peak at 30 days, parallel to its intracellular level. These results underline how metabolism changes during astrocyte differentiation, highlight that d-serine synthesis is restricted in differentiated astrocytes and provide a valuable model for developing potential novel therapeutic approaches to address brain diseases, especially the ones related to serine metabolism alterations.
Collapse
Affiliation(s)
- Farida Tripodi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Italy
| | - Zoraide Motta
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Giulia Murtas
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Valentina Rabattoni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Simona Nonnis
- DIVAS, Department of Veterinary Medicine and Animal Science, University of Milano, Italy
| | | | | | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy
- Department of Medical-Surgical Science and Biotechnologies, University of Rome La Sapienza, Italy
| | - Claudia Bearzi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy
- Institute for Biomedical Technologies, National Research Council of Italy (ITB-CNR), Milan, Italy
| | - Beatrice Badone
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Italy
| | - Silvia Sacchi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Gabriella Tedeschi
- DIVAS, Department of Veterinary Medicine and Animal Science, University of Milano, Italy
- CIMAINA, University of Milano, Italy
| | - Elisa Maffioli
- DIVAS, Department of Veterinary Medicine and Animal Science, University of Milano, Italy
| | - Paola Coccetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
28
|
Koh W, Kwak H, Cheong E, Lee CJ. GABA tone regulation and its cognitive functions in the brain. Nat Rev Neurosci 2023; 24:523-539. [PMID: 37495761 DOI: 10.1038/s41583-023-00724-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 07/28/2023]
Abstract
γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter released at GABAergic synapses, mediating fast-acting phasic inhibition. Emerging lines of evidence unequivocally indicate that a small amount of extracellular GABA - GABA tone - exists in the brain and induces a tonic GABA current that controls neuronal activity on a slow timescale relative to that of phasic inhibition. Surprisingly, studies indicate that glial cells that synthesize GABA, such as astrocytes, release GABA through non-vesicular mechanisms, such as channel-mediated release, and thereby act as the source of GABA tone in the brain. In this Review, we first provide an overview of major advances in our understanding of the cell-specific molecular and cellular mechanisms of GABA synthesis, release and clearance that regulate GABA tone in various brain regions. We next examine the diverse ways in which the tonic GABA current regulates synaptic transmission and synaptic plasticity through extrasynaptic GABAA-receptor-mediated mechanisms. Last, we discuss the physiological mechanisms through which tonic inhibition modulates cognitive function on a slow timescale. In this Review, we emphasize that the cognitive functions of tonic GABA current extend beyond mere inhibition, laying a foundation for future research on the physiological and pathophysiological roles of GABA tone regulation in normal and abnormal psychiatric conditions.
Collapse
Affiliation(s)
- Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea
| | - Hankyul Kwak
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea.
| |
Collapse
|
29
|
Çarçak N, Onat F, Sitnikova E. Astrocytes as a target for therapeutic strategies in epilepsy: current insights. Front Mol Neurosci 2023; 16:1183775. [PMID: 37583518 PMCID: PMC10423940 DOI: 10.3389/fnmol.2023.1183775] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/12/2023] [Indexed: 08/17/2023] Open
Abstract
Astrocytes are specialized non-neuronal glial cells of the central nervous system, contributing to neuronal excitability and synaptic transmission (gliotransmission). Astrocytes play a key roles in epileptogenesis and seizure generation. Epilepsy, as a chronic disorder characterized by neuronal hyperexcitation and hypersynchronization, is accompanied by substantial disturbances of glial cells and impairment of astrocytic functions and neuronal signaling. Anti-seizure drugs that provide symptomatic control of seizures primarily target neural activity. In epileptic patients with inadequate control of seizures with available anti-seizure drugs, novel therapeutic candidates are needed. These candidates should treat epilepsy with anti-epileptogenic and disease-modifying effects. Evidence from human and animal studies shows that astrocytes have value for developing new anti-seizure and anti-epileptogenic drugs. In this review, we present the key functions of astrocytes contributing to neuronal hyperexcitability and synaptic activity following an etiology-based approach. We analyze the role of astrocytes in both development (epileptogenesis) and generation of seizures (ictogenesis). Several promising new strategies that attempted to modify astroglial functions for treating epilepsy are being developed: (1) selective targeting of glia-related molecular mechanisms of glutamate transport; (2) modulation of tonic GABA release from astrocytes; (3) gliotransmission; (4) targeting the astrocytic Kir4.1-BDNF system; (5) astrocytic Na+/K+/ATPase activity; (6) targeting DNA hypo- or hypermethylation of candidate genes in astrocytes; (7) targeting astrocytic gap junction regulators; (8) targeting astrocytic adenosine kinase (the major adenosine-metabolizing enzyme); and (9) targeting microglia-astrocyte communication and inflammatory pathways. Novel disease-modifying therapeutic strategies have now been developed, such as astroglia-targeted gene therapy with a broad spectrum of genetic constructs to target astroglial cells.
Collapse
Affiliation(s)
- Nihan Çarçak
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey
- Institute of Health Sciences, Department of Neuroscience, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Filiz Onat
- Institute of Health Sciences, Department of Neuroscience, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Department of Medical Pharmacology, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Evgenia Sitnikova
- Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
30
|
Ma W, Si T, Wang Z, Wen P, Zhu Z, Liu Q, Wang J, Xu F, Li Q. Astrocytic α4-containing nAChR signaling in the hippocampus governs the formation of temporal association memory. Cell Rep 2023; 42:112674. [PMID: 37352098 DOI: 10.1016/j.celrep.2023.112674] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 03/24/2023] [Accepted: 06/06/2023] [Indexed: 06/25/2023] Open
Abstract
Everyday episodic memories involve linking together related events that are temporally separated. However, the mechanisms of forming this temporal association have remained unclear. Here, using astrocyte-specific manipulations, we show that potentiating astrocyte Ca2+ signaling in the hippocampal cornu ammonis 1 (CA1) enhances the strength of such temporal association, in parallel with long-term potentiation (LTP) enhancement of temporoammonic pathway to CA1, whereas attenuation of astrocyte Ca2+ signaling has the opposite effect. Moreover, we identify that these effects are mediated by astrocytic α4 subunit-containing nicotinic acetylcholine receptors (α4-nAChRs) via mechanisms involving NMDAR co-agonist supply. Finally, astrocytic α4-nAChRs underlie the cognitive enhancer nicotine's physiological effects. Together, these findings highlight the importance of astrocyte Ca2+ signaling in cognitive behavior and reveal a mechanism in governing the temporal association of episodic memory formation that operates through α4-nAChRs on hippocampal astrocytes.
Collapse
Affiliation(s)
- Wenyu Ma
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tengxiao Si
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zan Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pengjie Wen
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhenxiang Zhu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; Shenzhen Key Laboratory of Viral Vectors for Biomedicine, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Liu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fuqiang Xu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; Shenzhen Key Laboratory of Viral Vectors for Biomedicine, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qin Li
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
31
|
Yegla B, Rani A, Kumar A. Viral vector-mediated upregulation of serine racemase expression in medial prefrontal cortex improves learning and synaptic function in middle age rats. Aging (Albany NY) 2023; 15:2433-2449. [PMID: 37052995 PMCID: PMC10120901 DOI: 10.18632/aging.204652] [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: 01/19/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023]
Abstract
An age-associated decrease in N-methyl-D-aspartate receptor (NMDAR)-mediated synaptic function contributes to impaired synaptic plasticity and is associated with cognitive impairments. Levels of serine racemase (SR), an enzyme that synthesizes D-serine, an NMDAR co-agonist, decline with age. Thus, enhancing NMDAR function via increased SR expression in middle age, when subtle declines in cognition emerge, was predicted to enhance performance on a prefrontal cortex-mediated task sensitive to aging. Middle-aged (~12 mo) male Fischer-344 rats were injected bilaterally in the medial prefrontal cortex (mPFC) with viral vector (LV), SR (LV-SR) or control (LV-GFP). Rats were trained on the operant attentional set-shift task (AST) to examine cognitive flexibility and attentional function. LV-SR rats exhibited a faster rate of learning compared to controls during visual discrimination of the AST. Extradimensional set shifting and reversal were not impacted. Immunohistochemical analyses demonstrated that LV-SR significantly increased SR expression in the mPFC. Electrophysiological characterization of synaptic transmission in the mPFC slices obtained from LV-GFP and LV-SR animals indicated a significant increase in isolated NMDAR-mediated synaptic responses in LV-SR slices. Thus, results of the current study demonstrated that prefrontal SR upregulation in middle age rats can improve learning of task contingencies for visual discrimination and increase glutamatergic synaptic transmission, including NMDAR activity.
Collapse
Affiliation(s)
- Brittney Yegla
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
32
|
Astrocytes in memory formation and maintenance. Essays Biochem 2023; 67:107-117. [PMID: 36148596 DOI: 10.1042/ebc20220091] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022]
Abstract
Learning and memory are fundamental but highly complex functions of the brain. They rely on multiple mechanisms including the processing of sensory information, memory formation, maintenance of short- and long-term memory, memory retrieval and memory extinction. Recent experiments provide strong evidence that, besides neurons, astrocytes crucially contribute to these higher brain functions. However, the complex interplay of astrocytes and neurons in local neuron-glia assemblies is far from being understood. Although important basic cellular principles that govern and link neuronal and astrocytic cellular functions have been established, additional mechanisms clearly continue to emerge. In this short essay, we first review current technologies allowing the experimenter to explore the role of astrocytes in behaving animals, with focus on spatial memory. We then discuss astrocytic signaling mechanisms and their role in learning and memory. We also reveal gaps in our knowledge that currently prevent a comprehensive understanding of how astrocytes contribute to acquisition, storage and retrieval of memory by modulating neuronal signaling in local circuits.
Collapse
|
33
|
Goenaga J, Araque A, Kofuji P, Herrera Moro Chao D. Calcium signaling in astrocytes and gliotransmitter release. Front Synaptic Neurosci 2023; 15:1138577. [PMID: 36937570 PMCID: PMC10017551 DOI: 10.3389/fnsyn.2023.1138577] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Glia are as numerous in the brain as neurons and widely known to serve supportive roles such as structural scaffolding, extracellular ionic and neurotransmitter homeostasis, and metabolic support. However, over the past two decades, several lines of evidence indicate that astrocytes, which are a type of glia, play active roles in neural information processing. Astrocytes, although not electrically active, can exhibit a form of excitability by dynamic changes in intracellular calcium levels. They sense synaptic activity and release neuroactive substances, named gliotransmitters, that modulate neuronal activity and synaptic transmission in several brain areas, thus impacting animal behavior. This "dialogue" between astrocytes and neurons is embodied in the concept of the tripartite synapse that includes astrocytes as integral elements of synaptic function. Here, we review the recent work and discuss how astrocytes via calcium-mediated excitability modulate synaptic information processing at various spatial and time scales.
Collapse
Affiliation(s)
| | | | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
34
|
Liu Y, Shen X, Zhang Y, Zheng X, Cepeda C, Wang Y, Duan S, Tong X. Interactions of glial cells with neuronal synapses, from astrocytes to microglia and oligodendrocyte lineage cells. Glia 2023; 71:1383-1401. [PMID: 36799296 DOI: 10.1002/glia.24343] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 02/18/2023]
Abstract
The mammalian brain is a complex organ comprising neurons, glia, and more than 1 × 1014 synapses. Neurons are a heterogeneous group of electrically active cells, which form the framework of the complex circuitry of the brain. However, glial cells, which are primarily divided into astrocytes, microglia, oligodendrocytes (OLs), and oligodendrocyte precursor cells (OPCs), constitute approximately half of all neural cells in the mammalian central nervous system (CNS) and mainly provide nutrition and tropic support to neurons in the brain. In the last two decades, the concept of "tripartite synapses" has drawn great attention, which emphasizes that astrocytes are an integral part of the synapse and regulate neuronal activity in a feedback manner after receiving neuronal signals. Since then, synaptic modulation by glial cells has been extensively studied and substantially revised. In this review, we summarize the latest significant findings on how glial cells, in particular, microglia and OL lineage cells, impact and remodel the structure and function of synapses in the brain. Our review highlights the cellular and molecular aspects of neuron-glia crosstalk and provides additional information on how aberrant synaptic communication between neurons and glia may contribute to neural pathologies.
Collapse
Affiliation(s)
- Yao Liu
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Shen
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhan Zhang
- College of Basic Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoli Zheng
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Yao Wang
- Department of Assisted Reproduction, The Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shumin Duan
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China
| | - Xiaoping Tong
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| |
Collapse
|
35
|
Cuellar-Santoyo AO, Ruiz-Rodríguez VM, Mares-Barbosa TB, Patrón-Soberano A, Howe AG, Portales-Pérez DP, Miquelajáuregui Graf A, Estrada-Sánchez AM. Revealing the contribution of astrocytes to glutamatergic neuronal transmission. Front Cell Neurosci 2023; 16:1037641. [PMID: 36744061 PMCID: PMC9893894 DOI: 10.3389/fncel.2022.1037641] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Research on glutamatergic neurotransmission has focused mainly on the function of presynaptic and postsynaptic neurons, leaving astrocytes with a secondary role only to ensure successful neurotransmission. However, recent evidence indicates that astrocytes contribute actively and even regulate neuronal transmission at different levels. This review establishes a framework by comparing glutamatergic components between neurons and astrocytes to examine how astrocytes modulate or otherwise influence neuronal transmission. We have included the most recent findings about the role of astrocytes in neurotransmission, allowing us to understand the complex network of neuron-astrocyte interactions. However, despite the knowledge of synaptic modulation by astrocytes, their contribution to specific physiological and pathological conditions remains to be elucidated. A full understanding of the astrocyte's role in neuronal processing could open fruitful new frontiers in the development of therapeutic applications.
Collapse
Affiliation(s)
- Ares Orlando Cuellar-Santoyo
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Victor Manuel Ruiz-Rodríguez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Teresa Belem Mares-Barbosa
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | - Araceli Patrón-Soberano
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Andrew G. Howe
- Intelligent Systems Laboratory, HRL Laboratories, LLC, Malibu, CA, United States
| | - Diana Patricia Portales-Pérez
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | | | - Ana María Estrada-Sánchez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| |
Collapse
|
36
|
Carnosic Acid Attenuates AβOs-Induced Apoptosis and Synaptic Impairment via Regulating NMDAR2B and Its Downstream Cascades in SH-SY5Y Cells. Mol Neurobiol 2023; 60:133-144. [PMID: 36224322 DOI: 10.1007/s12035-022-03032-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 09/09/2022] [Indexed: 12/30/2022]
Abstract
Neuronal death and synaptic loss are principal pathological features of Alzheimer's disease (AD). Amyloid beta oligomers (AβOs) constitute the main neurotoxin underscoring AD pathology. AβOs interact with N-methyl-D-aspartate receptors (NMDARs), resulting in neurotoxic events, including activation of apoptosis and synaptic impairment. Carnosic acid (CA), extracted from Salvia rosmarinus, has been verified its neuroprotective effects in AD. However, the precise mechanisms by which CA induces synaptic protection remain unclear. In this study, we established an in vitro AD model using SH-SY5Y human neuroblastoma cells. We observed that CA improved neuronal survival by suppressing apoptosis. Moreover, CA restored synaptic impairments by increasing expression levels of brain-derived neurotrophic factor (BDNF), postsynaptic density protein-95 (PSD-95), and synaptophysin (Syn). Furthermore, we found these protective effects were dependent on inhibiting the phosphorylation of NMDAR subtype 2B (NMDAR2B), which further suppressed calcium overload and promoted activation of the extracellular signal-regulated kinase (ERK)-cAMP response element-binding protein (CREB) pathway. Administration of N-methyl-D-aspartic acid (NMDA), an agonist of NMDARs, abolished these effects of CA. Our findings demonstrate that CA exerts neuroprotective effects in an in vitro model of AD by regulating NMDAR2B and its downstream cascades, highlighting the therapeutic potential of CA as a NMDARs-targeted candidate in the treatment of AD.
Collapse
|
37
|
An astrocytic signaling loop for frequency-dependent control of dendritic integration and spatial learning. Nat Commun 2022; 13:7932. [PMID: 36566254 PMCID: PMC9789958 DOI: 10.1038/s41467-022-35620-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022] Open
Abstract
Dendrites of hippocampal CA1 pyramidal cells amplify clustered glutamatergic input by activation of voltage-gated sodium channels and N-methyl-D-aspartate receptors (NMDARs). NMDAR activity depends on the presence of NMDAR co-agonists such as D-serine, but how co-agonists influence dendritic integration is not well understood. Using combinations of whole-cell patch clamp, iontophoretic glutamate application, two-photon excitation fluorescence microscopy and glutamate uncaging in acute rat and mouse brain slices we found that exogenous D-serine reduced the threshold of dendritic spikes and increased their amplitude. Triggering an astrocytic mechanism controlling endogenous D-serine supply via endocannabinoid receptors (CBRs) also increased dendritic spiking. Unexpectedly, this pathway was activated by pyramidal cell activity primarily in the theta range, which required HCN channels and astrocytic CB1Rs. Therefore, astrocytes close a positive and frequency-dependent feedback loop between pyramidal cell activity and their integration of dendritic input. Its disruption in mice led to an impairment of spatial memory, which demonstrated its behavioral relevance.
Collapse
|
38
|
Kim S, Kwon J, Park MG, Lee CJ. Dopamine-induced astrocytic Ca 2+ signaling in mPFC is mediated by MAO-B in young mice, but by dopamine receptors in adult mice. Mol Brain 2022; 15:90. [PMID: 36397051 PMCID: PMC9670619 DOI: 10.1186/s13041-022-00977-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022] Open
Abstract
Dopamine (DA) plays a vital role in brain physiology and pathology such as learning and memory, motor control, neurological diseases, and psychiatric diseases. In neurons, it has been well established that DA increases or decreases intracellular cyclic AMP (cAMP) through D1-like or D2-like dopamine receptors, respectively. In contrast, it has been elusive how astrocytes respond to DA via Ca2+ signaling and regulate synaptic transmission and reward systems. Previous studies suggest various molecular targets such as MAO-B, D1R, or D1R-D2R heteromer to modulate astrocytic Ca2+ signaling. However, which molecular target is utilized under what physiological condition remains unclear. Here, we show that DA-induced astrocytic Ca2+ signaling pathway switches during development: MAO-B is the major player at a young age (5-6 weeks), whereas DA receptors (DARs) are responsible for the adult period (8-12 weeks). DA-mediated Ca2+ response in the adult period was decreased by either D1R or D2R blockers, which are primarily known for cyclic AMP signaling (Gs and Gi pathway, respectively), suggesting that this Ca2+ response might be mediated through Gq pathway by D1R-D2R heterodimer. Moreover, DAR-mediated Ca2+ response was not blocked by TTX, implying that this response is not a secondary response caused by neuronal activation. Our study proposes an age-specific molecular target of DA-induced astrocytic Ca2+ signaling: MAO-B in young mice and DAR in adult mice.
Collapse
Affiliation(s)
- Sunpil Kim
- grid.222754.40000 0001 0840 2678KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841 Republic of Korea ,grid.410720.00000 0004 1784 4496Center for Cognition and Sociality, Cognitive Glioscience Group, Institute for Basic Science (IBS), 55 Expo-Ro, Yusung-Gu, Daejeon, 34126 Republic of Korea
| | - Jea Kwon
- grid.410720.00000 0004 1784 4496Center for Cognition and Sociality, Cognitive Glioscience Group, Institute for Basic Science (IBS), 55 Expo-Ro, Yusung-Gu, Daejeon, 34126 Republic of Korea
| | - Mingu Gordon Park
- grid.222754.40000 0001 0840 2678KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841 Republic of Korea ,grid.410720.00000 0004 1784 4496Center for Cognition and Sociality, Cognitive Glioscience Group, Institute for Basic Science (IBS), 55 Expo-Ro, Yusung-Gu, Daejeon, 34126 Republic of Korea
| | - C. Justin Lee
- grid.222754.40000 0001 0840 2678KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841 Republic of Korea ,grid.410720.00000 0004 1784 4496Center for Cognition and Sociality, Cognitive Glioscience Group, Institute for Basic Science (IBS), 55 Expo-Ro, Yusung-Gu, Daejeon, 34126 Republic of Korea
| |
Collapse
|
39
|
Cho WH, Noh K, Lee BH, Barcelon E, Jun SB, Park HY, Lee SJ. Hippocampal astrocytes modulate anxiety-like behavior. Nat Commun 2022; 13:6536. [PMID: 36344520 PMCID: PMC9640657 DOI: 10.1038/s41467-022-34201-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/13/2022] [Indexed: 11/09/2022] Open
Abstract
Astrocytes can affect animal behavior by regulating tripartite synaptic transmission, yet their influence on affective behavior remains largely unclear. Here we showed that hippocampal astrocyte calcium activity reflects mouse affective state during virtual elevated plus maze test using two-photon calcium imaging in vivo. Furthermore, optogenetic hippocampal astrocyte activation elevating intracellular calcium induced anxiolytic behaviors in astrocyte-specific channelrhodopsin 2 (ChR2) transgenic mice (hGFAP-ChR2 mice). As underlying mechanisms, we found ATP released from the activated hippocampal astrocytes increased excitatory synaptic transmission in dentate gyrus (DG) granule cells, which exerted anxiolytic effects. Our data uncover a role of hippocampal astrocytes in modulating mice anxiety-like behaviors by regulating ATP-mediated synaptic homeostasis in hippocampal DG granule cells. Thus, manipulating hippocampal astrocytes activity can be a therapeutic strategy to treat anxiety.
Collapse
Affiliation(s)
- Woo-Hyun Cho
- grid.31501.360000 0004 0470 5905Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 08826 Republic of Korea
| | - Kyungchul Noh
- grid.31501.360000 0004 0470 5905Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 08826 Republic of Korea
| | - Byung Hun Lee
- grid.31501.360000 0004 0470 5905Department of Physics and Astronomy, Seoul National University, Seoul, 08826 Republic of Korea
| | - Ellane Barcelon
- grid.31501.360000 0004 0470 5905Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 08826 Republic of Korea
| | - Sang Beom Jun
- grid.255649.90000 0001 2171 7754Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, 03760 Republic of Korea ,grid.255649.90000 0001 2171 7754Graduate Program in Smart Factory, Ewha Womans University, Seoul, 03760 Republic of Korea ,grid.255649.90000 0001 2171 7754Department of Brain & Cognitive Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Hye Yoon Park
- grid.31501.360000 0004 0470 5905Department of Physics and Astronomy, Seoul National University, Seoul, 08826 Republic of Korea ,grid.17635.360000000419368657Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455 USA
| | - Sung Joong Lee
- grid.31501.360000 0004 0470 5905Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 08826 Republic of Korea
| |
Collapse
|
40
|
Owji AP, Yu K, Kittredge A, Wang J, Zhang Y, Yang T. Bestrophin-2 and glutamine synthetase form a complex for glutamate release. Nature 2022; 611:180-187. [PMID: 36289327 PMCID: PMC9873481 DOI: 10.1038/s41586-022-05373-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 09/21/2022] [Indexed: 01/26/2023]
Abstract
Bestrophin-2 (BEST2) is a member of the bestrophin family of calcium-activated anion channels that has a critical role in ocular physiology1-4. Here we uncover a directional permeability of BEST2 to glutamate that heavily favours glutamate exit, identify glutamine synthetase (GS) as a binding partner of BEST2 in the ciliary body of the eye, and solve the structure of the BEST2-GS complex. BEST2 reduces cytosolic GS activity by tethering GS to the cell membrane. GS extends the ion conducting pathway of BEST2 through its central cavity and inhibits BEST2 channel function in the absence of intracellular glutamate, but sensitizes BEST2 to intracellular glutamate, which promotes the opening of BEST2 and thus relieves the inhibitory effect of GS. We demonstrate the physiological role of BEST2 in conducting chloride and glutamate and the influence of GS in non-pigmented ciliary epithelial cells. Together, our results reveal a novel mechanism of glutamate release through BEST2-GS.
Collapse
Affiliation(s)
- Aaron P Owji
- Department of Ophthalmology, Columbia University, New York, NY, USA
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Kuai Yu
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, USA
| | - Alec Kittredge
- Department of Ophthalmology, Columbia University, New York, NY, USA
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Jiali Wang
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| | - Tingting Yang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| |
Collapse
|
41
|
Lee JM, Gadhe CG, Kang H, Pae AN, Lee CJ. Glutamate Permeability of Chicken Best1. Exp Neurobiol 2022; 31:277-288. [PMID: 36351838 PMCID: PMC9659495 DOI: 10.5607/en22038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 07/28/2023] Open
Abstract
Bestrophin-1 (Best1) is a calcium (Ca2+)-activated chloride (Cl-) channel which has a phylogenetically conserved channel structure with an aperture and neck in the ion-conducting pathway. Mammalian mouse Best1 (mBest1) has been known to have a permeability for large organic anions including gluconate, glutamate, and D-serine, in addition to several small monovalent anions, such as Cl‑, bromine (Br-), iodine (I-), and thiocyanate (SCN-). However, it is still unclear whether non-mammalian Best1 has a glutamate permeability through the ion-conducting pathway. Here, we report that chicken Best1 (cBest1) is permeable to glutamate in a Ca2+-dependent manner. The molecular docking and molecular dynamics simulation showed a glutamate binding at the aperture and neck of cBest1 and a glutamate permeation through the ion-conducting pore, respectively. Moreover, through electrophysiological recordings, we calculated the permeability ratio of glutamate to Cl- (PGlutamate/PCl) as 0.28 based on the reversal potential shift by ion substitution from Cl- to glutamate in the internal solution. Finally, we directly detected the Ca2+-dependent glutamate release through cBest1 using the ultrasensitive two-cell sniffer patch technique. Our results propose that Best1 homologs from non-mammalian (cBest1) to mammalian (mBest1) have a conserved permeability for glutamate.
Collapse
Affiliation(s)
- Jung Moo Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | | | - Hyunji Kang
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
- IBS School, University of Science and Technology, Daejeon 34113, Korea
| | - Ae Nim Pae
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
- KIST School, University of Science and Technology, Seoul 02792, Korea
| | - C. Justin Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
- IBS School, University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
42
|
Fuchsberger T, Paulsen O. Modulation of hippocampal plasticity in learning and memory. Curr Opin Neurobiol 2022; 75:102558. [PMID: 35660989 DOI: 10.1016/j.conb.2022.102558] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022]
Abstract
Synaptic plasticity plays a central role in the study of neural mechanisms of learning and memory. Plasticity rules are not invariant over time but are under neuromodulatory control, enabling behavioral states to influence memory formation. Neuromodulation controls synaptic plasticity at network level by directing information flow, at circuit level through changes in excitation/inhibition balance, and at synaptic level through modulation of intracellular signaling cascades. Although most research has focused on modulation of principal neurons, recent progress has uncovered important roles for interneurons in not only routing information, but also setting conditions for synaptic plasticity. Moreover, astrocytes have been shown to both gate and mediate plasticity. These additional mechanisms must be considered for a comprehensive mechanistic understanding of learning and memory.
Collapse
Affiliation(s)
- Tanja Fuchsberger
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
43
|
Nava-Gómez L, Calero-Vargas I, Higinio-Rodríguez F, Vázquez-Prieto B, Olivares-Moreno R, Ortiz-Retana J, Aranda P, Hernández-Chan N, Rojas-Piloni G, Alcauter S, López-Hidalgo M. AGING-ASSOCIATED COGNITIVE DECLINE IS REVERSED BY D-SERINE SUPPLEMENTATION. eNeuro 2022; 9:ENEURO.0176-22.2022. [PMID: 35584913 PMCID: PMC9186414 DOI: 10.1523/eneuro.0176-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 11/21/2022] Open
Abstract
Brain aging is a natural process that involves structural and functional changes that lead to cognitive decline, even in healthy subjects. This detriment has been associated with N-methyl-D-aspartate receptor (NMDAR) hypofunction due to a reduction in the brain levels of D-serine, the endogenous NMDAR co-agonist. However, it is not clear if D-serine supplementation could be used as an intervention to reduce or reverse age-related brain alterations. In the present work, we aimed to analyze the D-serine effect on aging-associated alterations in cellular and large-scale brain systems that could support cognitive flexibility in rats. We found that D-serine supplementation reverts the age-related decline in cognitive flexibility, frontal dendritic spine density, and partially restored large-scale functional connectivity without inducing nephrotoxicity; instead, D-serine restored the thickness of the renal epithelial cells that were affected by age. Our results suggest that D-serine could be used as a therapeutic target to reverse age-related brain alterations.SIGNIFICANT STATEMENTAge-related behavioral changes in cognitive performance occur as a physiological process of aging. Then, it is important to explore possible therapeutics to decrease, retard or reverse aging effects on the brain. NMDA receptor hypofunction contributes to the aging-associated cognitive decline. In the aged brain, there is a reduction in the brain levels of the NMDAR co-agonist, D-Serine. However, it is unclear if chronic D-serine supplementation could revert the age-detriment in brain functions. Our results show that D-serine supplementation reverts the age-associated decrease in cognitive flexibility, functional brain connectivity, and neuronal morphology. Our findings raise the possibility that restoring the brain levels of D-serine could be used as a therapeutic target to recover brain alterations associated with aging.
Collapse
Affiliation(s)
- L Nava-Gómez
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
- Facultad de Medicina. UAQ
| | - I Calero-Vargas
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - F Higinio-Rodríguez
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - B Vázquez-Prieto
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - R Olivares-Moreno
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - J Ortiz-Retana
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - P Aranda
- Facultad de Ciencias Naturales, UAQ
| | | | - G Rojas-Piloni
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - S Alcauter
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, México
| | - M López-Hidalgo
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla. UNAM
| |
Collapse
|
44
|
Berlin S. Astrocytes Get the BEST1 Out of NMDA Receptors. Biol Psychiatry 2022; 91:e29-e30. [PMID: 35331393 DOI: 10.1016/j.biopsych.2022.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Shai Berlin
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|