1
|
Verkhratsky A, Untiet V, Matchkov VV. Chloride fluxes and GABA release sustain inhibition in the CNS: The role for Bestrophin 1 anion channels. Acta Physiol (Oxf) 2025; 241:e14254. [PMID: 39587879 DOI: 10.1111/apha.14254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Neurosciences, University of the Basque Country, Leioa, Spain
- IKERBASQUE Basque Foundation for Science, Bilbao, Spain
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- International Joint Research Centre on Purinergic Signalling of Sichuan Province Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Verena Untiet
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
2
|
Di Palma M, Koh W, Lee CJ, Conti F. A quantitative analysis of bestrophin 1 cellular localization in mouse cerebral cortex. Acta Physiol (Oxf) 2025; 241:e14245. [PMID: 39466647 DOI: 10.1111/apha.14245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024]
Abstract
AIM Calcium-activated ligand-gated chloride channels, beyond their role in maintaining anion homeostasis, modulate neuronal excitability by facilitating nonvesicular neurotransmitter release. BEST1, a key member of this family, is permeable to γ-aminobutyric acid (GABA) and glutamate. While astrocytic BEST1 is well-studied and known to regulate neurotransmitter levels, its distribution and role in other brain cell types remain unclear. This study aimed to reassess the localization of BEST1 in the mouse cerebral cortex. METHODS We examined the localization and distribution of BEST1 in the mouse parietal cortex using light microscopy, confocal double-labeling with markers for astrocytes, neurons, microglia, and oligodendrocyte precursor cells, and 3D reconstruction techniques. RESULTS In the cerebral cortex, BEST1 is more broadly distributed than previously thought. Neurons are the second most abundant BEST1+ cell type in the cerebral cortex, following astrocytes. BEST1 is diffusely expressed in neuronal somatic and neuropilar domains and is present at glutamatergic and GABAergic terminals, with a prevalence at GABAergic terminals. We also confirmed that BEST1 is expressed in cortical microglia and identified it in oligodendrocyte precursor cells, albeit to a lesser extent. CONCLUSIONS Together, these findings suggest that BEST1's role in controlling neurotransmission may extend beyond astrocytes to include other brain cells. Understanding BEST1's function in these cells could offer new insights into the molecular mechanisms shaping cortical circuitry. Further research is needed to clarify the diverse roles of BEST1 in both normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Michael Di Palma
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| |
Collapse
|
3
|
Park J, Sipe GO, Tang X, Ojha P, Fernandes G, Leow YN, Zhang C, Osako Y, Natesan A, Drummond GT, Jaenisch R, Sur M. Astrocytic modulation of population encoding in mouse visual cortex via GABA transporter 3 revealed by multiplexed CRISPR/Cas9 gene editing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622321. [PMID: 39605420 PMCID: PMC11601534 DOI: 10.1101/2024.11.06.622321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Astrocytes, which are increasingly recognized as pivotal constituents of brain circuits governing a wide range of functions, express GABA transporter 3 (Gat3), an astrocyte-specific GABA transporter responsible for maintenance of extra-synaptic GABA levels. Here, we examined the functional role of Gat3 in astrocyte-mediated modulation of neuronal activity and information encoding. First, we developed a multiplexed CRISPR construct applicable for effective genetic ablation of Gat3 in the visual cortex of adult mice. Using in vivo two-photon calcium imaging of visual cortex neurons in Gat3 knockout mice, we observed changes in spontaneous and visually driven single neuronal response properties such as response magnitudes and trial-to-trial variability. Gat3 knockout exerted a pronounced influence on population-level neuronal activity, altering the response dynamics of neuronal populations and impairing their ability to accurately represent stimulus information. These findings demonstrate that Gat3 in astrocytes profoundly shapes the sensory information encoding capacity of neurons and networks within the visual cortex.
Collapse
Affiliation(s)
- Jiho Park
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- These authors contributed equally
| | - Grayson O. Sipe
- Department of Biology, Eberly College of Science and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
- These authors contributed equally
| | - Xin Tang
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Prachi Ojha
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Giselle Fernandes
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Yi Ning Leow
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Caroline Zhang
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
| | - Yuma Osako
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Arundhati Natesan
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Gabrielle T. Drummond
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
4
|
Sutley-Koury SN, Anderson A, Taitano-Johnson C, Ajayi M, Kulinich AO, Contreras K, Regalado J, Tiwari-Woodruff SK, Ethell IM. Astrocytic Ephrin-B1 Regulates Oligodendrocyte Development and Myelination. ASN Neuro 2024; 16:2401753. [PMID: 39437409 DOI: 10.1080/17590914.2024.2401753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Abstract
Astrocytes have been implicated in oligodendrocyte development and myelination, however, the mechanisms by which astrocytes regulate oligodendrocytes remain unclear. Our findings suggest a new mechanism that regulates astrocyte-mediated oligodendrocyte development through ephrin-B1 signaling in astrocytes. Using a mouse model, we examined the role of astrocytic ephrin-B1 signaling in oligodendrocyte development by deleting ephrin-B1 specifically in astrocytes during the postnatal days (P)14-P28 period and used mRNA analysis, immunohistochemistry, and mouse behaviors to study its effects on oligodendrocytes and myelination. We found that deletion of astrocytic ephrin-B1 downregulated many genes associated with oligodendrocyte development, myelination, and lipid metabolism in the hippocampus and the corpus callosum. Additionally, we observed a reduced number of oligodendrocytes and impaired myelination in the corpus callosum of astrocyte-specific ephrin-B1 KO mice. Finally, our data show reduced motor strength in these mice exhibiting clasping phenotype and impaired performance in the rotarod test most likely due to impaired myelination. Our studies provide new evidence that astrocytic ephrin-B1 positively regulates oligodendrocyte development and myelination, potentially through astrocyte-oligodendrocyte interactions.
Collapse
Affiliation(s)
- Samantha N Sutley-Koury
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Alyssa Anderson
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Christopher Taitano-Johnson
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
- Neuroscience Graduate Program, University of California Riverside, Riverside, California, USA
| | - Moyinoluwa Ajayi
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Anna O Kulinich
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Kimberly Contreras
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Jasmin Regalado
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
- Neuroscience Graduate Program, University of California Riverside, Riverside, California, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
- Neuroscience Graduate Program, University of California Riverside, Riverside, California, USA
| |
Collapse
|
5
|
Wang J, Owji AP, Kittredge A, Clark Z, Zhang Y, Yang T. GAD65 tunes the functions of Best1 as a GABA receptor and a neurotransmitter conducting channel. Nat Commun 2024; 15:8051. [PMID: 39277606 PMCID: PMC11401937 DOI: 10.1038/s41467-024-52039-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 08/23/2024] [Indexed: 09/17/2024] Open
Abstract
Bestrophin-1 (Best1) is an anion channel genetically linked to vision-threatening retinal degenerative channelopathies. Here, we identify interactions between Best1 and both isoforms of glutamic acid decarboxylases (GAD65 and GAD67), elucidate the distinctive influences of GAD65 and GAD67 on Best1's permeability to various anions/neurotransmitters, discover the functionality of Best1 as a γ-Aminobutyric acid (GABA) type A receptor, and solve the structure of GABA-bound Best1. GAD65 and GAD67 both promote Best1-mediated Cl- currents, but only GAD65 drastically enhances the permeability of Best1 to glutamate and GABA, for which GAD67 has no effect. GABA binds to Best1 on an extracellular site and stimulates Best1-mediated Cl- currents at the nano-molar concentration level. The physiological role of GAD65 as a cell type-specific binding partner and facilitator of Best1 is demonstrated in retinal pigment epithelial cells. Together, our results reveal critical regulators of Best1 and inform a network of membrane transport metabolons formed between bestrophin channels and glutamate metabolic enzymes.
Collapse
Affiliation(s)
- Jiali Wang
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Aaron P Owji
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Alec Kittredge
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Zada Clark
- 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
|
6
|
Wang Y, Wang Z. Effects and Safety of Monoamine Oxidase-B Inhibitors for Early Parkinson's Disease: A Network Meta-Analysis. Eur Neurol 2024; 87:273-290. [PMID: 39278214 DOI: 10.1159/000541315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/02/2024] [Indexed: 09/18/2024]
Abstract
INTRODUCTION The objective of this study was to evaluate the effects and safety of monoamine oxidase-B inhibitors (MAO-B inhibitors) for early Parkinson's disease (PD). METHODS All studies that assessed the efficacy of MAO-B inhibitors in patients with early PD were searched. Publications were screened, and data were extracted according to predefined criteria. Rev Man 5.4 and Stata 14.0 software were used for statistical analysis. Outcomes assessed included change of Unified Parkinson's Disease Rating Scale (UPDRS) total score, UPDRS part II score, UPDRS part III score, and the incidence of adverse events. RESULTS Thirty trials were identified and included in this meta-analysis. Compared with placebo, rasagiline, selegiline, safinamide, and zonisamide were significantly more effective, with a standardized mean difference (SMD) of -0.41 (95% confidence interval (CI) = -0.64 to -0.18), SMD = -0.38 (95% CI = -0.51 to -0.24), SMD = -0.37 (95% CI = -0.54 to -0.21), and SMD = -0.31 (95% CI = -0.57 to -0.05) on the UPDRS III score change, respectively. The surface under the cumulative ranking results showed that rasagiline ranked first in improving UPDRS II and UPDRS III, respectively. For safety outcomes, safinamide combination with dopaminergic treatment had lower risk of incurring any adverse events (risk ratio = 0.1, 95% CI = 0.01-0.2), and no statistical difference in incidence of adverse events was observed among other MAO-B inhibitor regimes and placebo. CONCLUSION Rasagiline, selegiline, safinamide, and zonisamide were effective compared to placebo in the treatment of early PD, but rasagiline was the most effective drug. As for safety, safinamide combination with dopaminergic treatment had lower risk of incurring any adverse events.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| | - Zhiyun Wang
- Department of Neurology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
7
|
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
|
8
|
Hayashi Y, Alamir N, Sun G, Tamagnini F, Hayashi Y, Williams C, Zheng Y. An effective textured Novel Object Recognition Test (tNORT) for repeated measure of whisker sensitivity of rodents. Behav Brain Res 2024; 472:115153. [PMID: 39025432 DOI: 10.1016/j.bbr.2024.115153] [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: 03/20/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Rodents use their whisker system to discriminate surface texture. Whisker-based texture discrimination tasks are often used to investigate the mechanisms encoding tactile sensation. One such task is the textured Novel Object Recognition Test (tNORT). It takes advantage of a tendency of rodents to explore novel objects more than familiar ones and assesses the sensitivity of whiskers in discriminating different textures of objects. It requires little training of the animals and the equipment involved is a simple arena with typically two objects placed inside. The success of the test relies on rodents spending sufficient time exploring these objects. Animals may lose interests in such tasks when performed repetitively within a limited time frame. However, such repeated tests may be crucial when establishing a sensitivity threshold of the whisker system. Here we present an adapted rodent tNORT protocol designed to maintain sustained interest in the objects even with repeated testing. We constructed complex objects from three simple-shaped objects. Different textures were provided by sandpapers of varying grit sizes. To minimise olfactory clues, we used the sandy and the laminar side of the same sandpaper as the familiar and novel textures assigned at random. We subsequently conducted repeated tNORTs on eight rats in order to identify a critical threshold of the sandpaper grit size below which rats would be unable to discriminate the sandy from the laminar side. With an inter-test-interval of seven days and after five tNORTs, the protocol enabled us to successfully identify the threshold. We suggest that the proposed tNORT is a useful tool for investigating the sensitivity threshold of the whisker system of rodent, and for testing the effectiveness of an intervention by comparing sensitivity threshold pre- and post-intervention.
Collapse
Affiliation(s)
- Yurie Hayashi
- School of Biological Sciences, Whiteknights, University of Reading, Reading RG6 7AY, UK
| | - Najeeba Alamir
- School of Biological Sciences, Whiteknights, University of Reading, Reading RG6 7AY, UK
| | - Guoyang Sun
- School of Biological Sciences, Whiteknights, University of Reading, Reading RG6 7AY, UK
| | - Francesco Tamagnini
- School of Pharmacy, University of Reading, Whiteknights, Reading RG6 6LA, UK; Centre for Integrative Neuroscience and Neurodynamics (CINN), University of Reading, Reading RG6 6AL, UK
| | - Yoshikatsu Hayashi
- School of Biological Sciences, Whiteknights, University of Reading, Reading RG6 7AY, UK
| | - Claire Williams
- Centre for Integrative Neuroscience and Neurodynamics (CINN), University of Reading, Reading RG6 6AL, UK; School of Psychology and Clinical Language Science, Whiteknights, University of Reading, Reading RG6 6AL, UK
| | - Ying Zheng
- School of Biological Sciences, Whiteknights, University of Reading, Reading RG6 7AY, UK; Centre for Integrative Neuroscience and Neurodynamics (CINN), University of Reading, Reading RG6 6AL, UK.
| |
Collapse
|
9
|
Shigetomi E, Suzuki H, Hirayama YJ, Sano F, Nagai Y, Yoshihara K, Koga K, Tateoka T, Yoshioka H, Shinozaki Y, Kinouchi H, Tanaka KF, Bito H, Tsuda M, Koizumi S. Disease-relevant upregulation of P2Y 1 receptor in astrocytes enhances neuronal excitability via IGFBP2. Nat Commun 2024; 15:6525. [PMID: 39117630 PMCID: PMC11310333 DOI: 10.1038/s41467-024-50190-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 06/26/2024] [Indexed: 08/10/2024] Open
Abstract
Reactive astrocytes play a pivotal role in the pathogenesis of neurological diseases; however, their functional phenotype and the downstream molecules by which they modify disease pathogenesis remain unclear. Here, we genetically increase P2Y1 receptor (P2Y1R) expression, which is upregulated in reactive astrocytes in several neurological diseases, in astrocytes of male mice to explore its function and the downstream molecule. This astrocyte-specific P2Y1R overexpression causes neuronal hyperexcitability by increasing both astrocytic and neuronal Ca2+ signals. We identify insulin-like growth factor-binding protein 2 (IGFBP2) as a downstream molecule of P2Y1R in astrocytes; IGFBP2 acts as an excitatory signal to cause neuronal excitation. In neurological disease models of epilepsy and stroke, reactive astrocytes upregulate P2Y1R and increase IGFBP2. The present findings identify a mechanism underlying astrocyte-driven neuronal hyperexcitability, which is likely to be shared by several neurological disorders, providing insights that might be relevant for intervention in diverse neurological disorders.
Collapse
Affiliation(s)
- Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan.
| | - Hideaki Suzuki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Yukiho J Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Fumikazu Sano
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Yuki Nagai
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Kohei Yoshihara
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keisuke Koga
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Neurophysiology, Hyogo College of Medicine, Hyogo, 663-8501, Japan
| | - Toru Tateoka
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hideyuki Yoshioka
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hiroyuki Kinouchi
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan.
| |
Collapse
|
10
|
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 2024:10.1007/s11302-024-10034-x. [PMID: 39046648 DOI: 10.1007/s11302-024-10034-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [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
|
11
|
Kang H, Han AR, Zhang A, Jeong H, Koh W, Lee JM, Lee H, Jo HY, Maria-Solano MA, Bhalla M, Kwon J, Roh WS, Yang J, An HJ, Choi S, Kim HM, Lee CJ. GolpHCat (TMEM87A), a unique voltage-dependent cation channel in Golgi apparatus, contributes to Golgi-pH maintenance and hippocampus-dependent memory. Nat Commun 2024; 15:5830. [PMID: 38992057 PMCID: PMC11239671 DOI: 10.1038/s41467-024-49297-8] [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: 12/14/2023] [Accepted: 05/30/2024] [Indexed: 07/13/2024] Open
Abstract
Impaired ion channels regulating Golgi pH lead to structural alterations in the Golgi apparatus, such as fragmentation, which is found, along with cognitive impairment, in Alzheimer's disease. However, the causal relationship between altered Golgi structure and cognitive impairment remains elusive due to the lack of understanding of ion channels in the Golgi apparatus of brain cells. Here, we identify that a transmembrane protein TMEM87A, renamed Golgi-pH-regulating cation channel (GolpHCat), expressed in astrocytes and neurons that contributes to hippocampus-dependent memory. We find that GolpHCat displays unique voltage-dependent currents, which is potently inhibited by gluconate. Additionally, we gain structural insights into the ion conduction through GolpHCat at the molecular level by determining three high-resolution cryogenic-electron microscopy structures of human GolpHCat. GolpHCat-knockout mice show fragmented Golgi morphology and altered protein glycosylation and functions in the hippocampus, leading to impaired spatial memory. These findings suggest a molecular target for Golgi-related diseases and cognitive impairment.
Collapse
Affiliation(s)
- Hyunji Kang
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea
- IBS School, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Ah-Reum Han
- Center for Biomolecular and Cellular Structure, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea
| | - Aihua Zhang
- Global AI Drug Discovery Center, College of Pharmacy and Graduate School of Pharmaceutical Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Heejin Jeong
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea
| | - Jung Moo Lee
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea
| | - Hayeon Lee
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea
| | - Hee Young Jo
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Korea
| | - Miguel A Maria-Solano
- Global AI Drug Discovery Center, College of Pharmacy and Graduate School of Pharmaceutical Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Mridula Bhalla
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea
| | - Jea Kwon
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea
| | - Woo Suk Roh
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea
| | - Jimin Yang
- Center for Biomolecular and Cellular Structure, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea
| | - Hyun Joo An
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Korea
| | - Sun Choi
- Global AI Drug Discovery Center, College of Pharmacy and Graduate School of Pharmaceutical Science, Ewha Womans University, Seoul, 03760, Republic of Korea.
| | - Ho Min Kim
- Center for Biomolecular and Cellular Structure, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea.
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| | - C Justin Lee
- Center for Cognition and Sociality, Life Science Cluster, Institute for Basic Science (IBS), 55 Expo-ro, Yuseong-gu, Daejeon, 34126, Republic of Korea.
- IBS School, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
12
|
Chen K, Forrest AM, Burgos GG, Kozai TDY. Neuronal functional connectivity is impaired in a layer dependent manner near chronically implanted intracortical microelectrodes in C57BL6 wildtype mice. J Neural Eng 2024; 21:036033. [PMID: 38788704 DOI: 10.1088/1741-2552/ad5049] [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: 05/24/2024] [Indexed: 05/26/2024]
Abstract
Objective.This study aims to reveal longitudinal changes in functional network connectivity within and across different brain structures near chronically implanted microelectrodes. While it is well established that the foreign-body response (FBR) contributes to the gradual decline of the signals recorded from brain implants over time, how the FBR affects the functional stability of neural circuits near implanted brain-computer interfaces (BCIs) remains unknown. This research aims to illuminate how the chronic FBR can alter local neural circuit function and the implications for BCI decoders.Approach.This study utilized single-shank, 16-channel,100µm site-spacing Michigan-style microelectrodes (3 mm length, 703µm2 site area) that span all cortical layers and the hippocampal CA1 region. Sex balanced C57BL6 wildtype mice (11-13 weeks old) received perpendicularly implanted microelectrode in left primary visual cortex. Electrophysiological recordings were performed during both spontaneous activity and visual sensory stimulation. Alterations in neuronal activity near the microelectrode were tested assessing cross-frequency synchronization of local field potential (LFP) and spike entrainment to LFP oscillatory activity throughout 16 weeks after microelectrode implantation.Main results. The study found that cortical layer 4, the input-receiving layer, maintained activity over the implantation time. However, layers 2/3 rapidly experienced severe impairment, leading to a loss of proper intralaminar connectivity in the downstream output layers 5/6. Furthermore, the impairment of interlaminar connectivity near the microelectrode was unidirectional, showing decreased connectivity from Layers 2/3 to Layers 5/6 but not the reverse direction. In the hippocampus, CA1 neurons gradually became unable to properly entrain to the surrounding LFP oscillations.Significance. This study provides a detailed characterization of network connectivity dysfunction over long-term microelectrode implantation periods. This new knowledge could contribute to the development of targeted therapeutic strategies aimed at improving the health of the tissue surrounding brain implants and potentially inform engineering of adaptive decoders as the FBR progresses. Our study's understanding of the dynamic changes in the functional network over time opens the door to developing interventions for improving the long-term stability and performance of intracortical microelectrodes.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Adam M Forrest
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | | | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
13
|
Graziano B, Wang L, White OR, Kaplan DH, Fernandez-Abascal J, Bianchi L. Glial KCNQ K + channels control neuronal output by regulating GABA release from glia in C. elegans. Neuron 2024; 112:1832-1847.e7. [PMID: 38460523 PMCID: PMC11156561 DOI: 10.1016/j.neuron.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/22/2024] [Accepted: 02/16/2024] [Indexed: 03/11/2024]
Abstract
KCNQs are voltage-gated K+ channels that control neuronal excitability and are mutated in epilepsy and autism spectrum disorder (ASD). KCNQs have been extensively studied in neurons, but their function in glia is unknown. Using voltage, calcium, and GABA imaging, optogenetics, and behavioral assays, we show here for the first time in Caenorhabditis elegans (C. elegans) that glial KCNQ channels control neuronal excitability by mediating GABA release from glia via regulation of the function of L-type voltage-gated Ca2+ channels. Further, we show that human KCNQ channels have the same role when expressed in nematode glia, underscoring conservation of function across species. Finally, we show that pathogenic loss-of-function and gain-of-function human KCNQ2 mutations alter glia-to-neuron GABA signaling in distinct ways and that the KCNQ channel opener retigabine exerts rescuing effects. This work identifies glial KCNQ channels as key regulators of neuronal excitability via control of GABA release from glia.
Collapse
Affiliation(s)
- Bianca Graziano
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lei Wang
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Olivia R White
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daryn H Kaplan
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jesus Fernandez-Abascal
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Laura Bianchi
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
14
|
Won D, Lee EH, Chang JE, Nam MH, Park KD, Oh SJ, Hwang JY. The role of astrocytic γ-aminobutyric acid in the action of inhalational anesthetics. Eur J Pharmacol 2024; 970:176494. [PMID: 38484926 DOI: 10.1016/j.ejphar.2024.176494] [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: 12/04/2023] [Revised: 01/24/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Inhalational anesthetics target the inhibitory extrasynaptic γ-aminobutyric acid type A (GABAA) receptors. Both neuronal and glial GABA mediate tonic inhibition of the extrasynaptic GABAA receptors. However, the role of glial GABA during inhalational anesthesia remains unclear. This study aimed to evaluate whether astrocytic GABA contributes to the action of different inhalational anesthetics. METHODS Gene knockout of monoamine oxidase B (MAOB) was used to reduce astrocytic GABA levels in mice. The hypnotic and immobilizing effects of isoflurane, sevoflurane, and desflurane were assessed by evaluating the loss of righting reflex (LORR) and tail-pinch withdrawal response (LTWR) in MAOB knockout and wild-type mice. Minimum alveolar concentration (MAC) for LORR, time to LORR, MAC for LTWR and time to LTWR of isoflurane, sevoflurane, and desflurane were assessed. RESULTS Time to LORR and time to LTWR with isoflurane were significantly longer in MAOB knockout mice than in wild-type mice (P < 0.001 and P = 0.032, respectively). Time to LORR with 0.8 MAC of sevoflurane was significantly longer in MAOB knockout mice than in wild-type mice (P < 0.001), but not with 1.0 MAC of sevoflurane (P=0.217). MAC for LTWR was significantly higher in MAOB knockout mice exposed to sevoflurane (P < 0.001). With desflurane, MAOB knockout mice had a significantly higher MAC for LORR (P = 0.003) and higher MAC for LTWR (P < 0.001) than wild-type mice. CONCLUSIONS MAOB knockout mice showed reduced sensitivity to the hypnotic and immobilizing effects of isoflurane, sevoflurane, and desflurane. Behavioral tests revealed that the hypnotic and immobilizing effects of inhalational anesthetics would be mediated by astrocytic GABA.
Collapse
Affiliation(s)
- Dongwook Won
- Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea; College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Elliot H Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jee-Eun Chang
- Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea; College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ki Duk Park
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.
| | - Jin-Young Hwang
- Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea; College of Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Rothman DL, Behar KL, Dienel GA. Mechanistic stoichiometric relationship between the rates of neurotransmission and neuronal glucose oxidation: Reevaluation of and alternatives to the pseudo-malate-aspartate shuttle model. J Neurochem 2024; 168:555-591. [PMID: 36089566 DOI: 10.1111/jnc.15619] [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: 11/12/2021] [Revised: 04/08/2022] [Accepted: 04/15/2022] [Indexed: 11/26/2022]
Abstract
The ~1:1 stoichiometry between the rates of neuronal glucose oxidation (CMRglc-ox-N) and glutamate (Glu)/γ-aminobutyric acid (GABA)-glutamine (Gln) neurotransmitter (NT) cycling between neurons and astrocytes (VNTcycle) has been firmly established. However, the mechanistic basis for this relationship is not fully understood, and this knowledge is critical for the interpretation of metabolic and brain imaging studies in normal and diseased brain. The pseudo-malate-aspartate shuttle (pseudo-MAS) model established the requirement for glycolytic metabolism in cultured glutamatergic neurons to produce NADH that is shuttled into mitochondria to support conversion of extracellular Gln (i.e., astrocyte-derived Gln in vivo) into vesicular neurotransmitter Glu. The evaluation of this model revealed that it could explain half of the 1:1 stoichiometry and it has limitations. Modifications of the pseudo-MAS model were, therefore, devised to address major knowledge gaps, that is, submitochondrial glutaminase location, identities of mitochondrial carriers for Gln and other model components, alternative mechanisms to transaminate α-ketoglutarate to form Glu and shuttle glutamine-derived ammonia while maintaining mass balance. All modified models had a similar 0.5 to 1.0 predicted mechanistic stoichiometry between VNTcycle and the rate of glucose oxidation. Based on studies of brain β-hydroxybutyrate oxidation, about half of CMRglc-ox-N may be linked to glutamatergic neurotransmission and localized in pre-synaptic structures that use pseudo-MAS type mechanisms for Glu-Gln cycling. In contrast, neuronal compartments that do not participate in transmitter cycling may use the MAS to sustain glucose oxidation. The evaluation of subcellular compartmentation of neuronal glucose metabolism in vivo is a critically important topic for future studies to understand glutamatergic and GABAergic neurotransmission.
Collapse
Affiliation(s)
- Douglas L Rothman
- Magnetic Resonance Research Center and Departments of Radiology and Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Kevin L Behar
- Magnetic Resonance Research Center and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| |
Collapse
|
16
|
Djama D, Zirpel F, Ye Z, Moore G, Chue C, Edge C, Jager P, Delogu A, Brickley SG. The type of inhibition provided by thalamic interneurons alters the input selectivity of thalamocortical neurons. CURRENT RESEARCH IN NEUROBIOLOGY 2024; 6:100130. [PMID: 38694514 PMCID: PMC11061260 DOI: 10.1016/j.crneur.2024.100130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
A fundamental problem in neuroscience is how neurons select for their many inputs. A common assumption is that a neuron's selectivity is largely explained by differences in excitatory synaptic input weightings. Here we describe another solution to this important problem. We show that within the first order visual thalamus, the type of inhibition provided by thalamic interneurons has the potential to alter the input selectivity of thalamocortical neurons. To do this, we developed conductance injection protocols to compare how different types of synchronous and asynchronous GABA release influence thalamocortical excitability in response to realistic patterns of retinal ganglion cell input. We show that the asynchronous GABA release associated with tonic inhibition is particularly efficient at maintaining information content, ensuring that thalamocortical neurons can distinguish between their inputs. We propose a model where alterations in GABA release properties results in rapid changes in input selectivity without requiring structural changes in the network.
Collapse
Affiliation(s)
- Deyl Djama
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Florian Zirpel
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Zhiwen Ye
- Department of Biological Structure, University of Washington, Seattle, USA
| | - Gerald Moore
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Charmaine Chue
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Christopher Edge
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Polona Jager
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9NU, UK
| | - Alessio Delogu
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9NU, UK
| | | |
Collapse
|
17
|
Rohner VL, Lamothe-Molina PJ, Patriarchi T. Engineering, applications, and future perspectives of GPCR-based genetically encoded fluorescent indicators for neuromodulators. J Neurochem 2024; 168:163-184. [PMID: 38288673 DOI: 10.1111/jnc.16045] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 02/23/2024]
Abstract
This review explores the evolving landscape of G-protein-coupled receptor (GPCR)-based genetically encoded fluorescent indicators (GEFIs), with a focus on their development, structural components, engineering strategies, and applications. We highlight the unique features of this indicator class, emphasizing the importance of both the sensing domain (GPCR structure and activation mechanism) and the reporting domain (circularly permuted fluorescent protein (cpFP) structure and fluorescence modulation). Further, we discuss indicator engineering approaches, including the selection of suitable cpFPs and expression systems. Additionally, we showcase the diversity and flexibility of their application by presenting a summary of studies where such indicators were used. Along with all the advantages, we also focus on the current limitations as well as common misconceptions that arise when using these indicators. Finally, we discuss future directions in indicator engineering, including strategies for screening with increased throughput, optimization of the ligand-binding properties, structural insights, and spectral diversity.
Collapse
Affiliation(s)
- Valentin Lu Rohner
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
| | | | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Benarroch E. What Is the Role of the "GABA Tone" in Normal and Pathological Conditions? Neurology 2024; 102:e209152. [PMID: 38252909 DOI: 10.1212/wnl.0000000000209152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 11/28/2023] [Indexed: 01/24/2024] Open
|
19
|
Ahmadpour N, Kantroo M, Stobart MJ, Meza-Resillas J, Shabanipour S, Parra-Nuñez J, Salamovska T, Muzaleva A, O'Hara F, Erickson D, Di Gaetano B, Carrion-Falgarona S, Weber B, Lamont A, Lavine NE, Kauppinen TM, Jackson MF, Stobart JL. Cortical astrocyte N-methyl-D-aspartate receptors influence whisker barrel activity and sensory discrimination in mice. Nat Commun 2024; 15:1571. [PMID: 38383567 PMCID: PMC10882001 DOI: 10.1038/s41467-024-45989-3] [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: 01/08/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024] Open
Abstract
Astrocytes express ionotropic receptors, including N-methyl-D-aspartate receptors (NMDARs). However, the contribution of NMDARs to astrocyte-neuron interactions, particularly in vivo, has not been elucidated. Here we show that a knockdown approach to selectively reduce NMDARs in mouse cortical astrocytes decreases astrocyte Ca2+ transients evoked by sensory stimulation. Astrocyte NMDAR knockdown also impairs nearby neuronal circuits by elevating spontaneous neuron activity and limiting neuronal recruitment, synchronization, and adaptation during sensory stimulation. Furthermore, this compromises the optimal processing of sensory information since the sensory acuity of the mice is reduced during a whisker-dependent tactile discrimination task. Lastly, we rescue the effects of astrocyte NMDAR knockdown on neurons and improve the tactile acuity of the animal by supplying exogenous ATP. Overall, our findings show that astrocytes can respond to nearby neuronal activity via their NMDAR, and that these receptors are an important component for purinergic signaling that regulate astrocyte-neuron interactions and cortical sensory discrimination in vivo.
Collapse
Affiliation(s)
| | - Meher Kantroo
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | | | | | | | | | | | - Anna Muzaleva
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | - Finnegan O'Hara
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | - Dustin Erickson
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | - Bruno Di Gaetano
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | | | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Alana Lamont
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- PrairieNeuro Research Center, Health Sciences Center, Winnipeg, MB, Canada
| | - Natalie E Lavine
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- PrairieNeuro Research Center, Health Sciences Center, Winnipeg, MB, Canada
| | - Tiina M Kauppinen
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- PrairieNeuro Research Center, Health Sciences Center, Winnipeg, MB, Canada
| | - Michael F Jackson
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
- PrairieNeuro Research Center, Health Sciences Center, Winnipeg, MB, Canada
| | - Jillian L Stobart
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada.
- Centre on Aging, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
20
|
Ali DN, Ali HM, Lopez MR, Kang S, Choi DS. Astrocytic GABAergic Regulation in Alcohol Use and Major Depressive Disorders. Cells 2024; 13:318. [PMID: 38391931 PMCID: PMC10887002 DOI: 10.3390/cells13040318] [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: 11/30/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system (CNS). Most GABAergic neurons synthesize GABA from glutamate and release it in the synaptic cleft in the CNS. However, astrocytes can also synthesize and release GABA, activating GABA receptors in the neighboring neurons in physiological and pathological conditions. As the primary homeostatic glial cells in the brain, astrocytes play a crucial role in regulating GABA homeostasis and synaptic neurotransmission. Accumulating evidence demonstrates that astrocytic GABA dysregulation is implicated in psychiatric disorders, including alcohol use disorder (AUD) and major depressive disorder (MDD), the most prevalent co-occurring psychiatric disorders. Several current medications and emerging pharmacological agents targeting GABA levels are in clinical trials for treating AUD and MDD. This review offers a concise summary of the role of astrocytic GABA regulation in AUD and MDD. We also provide an overview of the current understanding and areas of debate regarding the mechanisms by which astrocytes regulate GABA in the CNS and their potential significance in the molecular basis of AUD and MDD, paving the way toward future research directions and potential therapeutic target areas within this field.
Collapse
Affiliation(s)
- Dina N. Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Hossam M. Ali
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Matthew R. Lopez
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Shinwoo Kang
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
| | - Doo-Sup Choi
- Department of Molecular Pharmacology and Experimental Therapeutics, Rochester, MN 55905, USA; (D.N.A.); (H.M.A.); (M.R.L.); (S.K.)
- Neuroscience Program, Rochester, MN 55905, USA
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
21
|
Tempone MH, Borges-Martins VP, César F, Alexandrino-Mattos DP, de Figueiredo CS, Raony Í, dos Santos AA, Duarte-Silva AT, Dias MS, Freitas HR, de Araújo EG, Ribeiro-Resende VT, Cossenza M, P. Silva H, P. de Carvalho R, Ventura ALM, Calaza KC, Silveira MS, Kubrusly RCC, de Melo Reis RA. The Healthy and Diseased Retina Seen through Neuron-Glia Interactions. Int J Mol Sci 2024; 25:1120. [PMID: 38256192 PMCID: PMC10817105 DOI: 10.3390/ijms25021120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
The retina is the sensory tissue responsible for the first stages of visual processing, with a conserved anatomy and functional architecture among vertebrates. To date, retinal eye diseases, such as diabetic retinopathy, age-related macular degeneration, retinitis pigmentosa, glaucoma, and others, affect nearly 170 million people worldwide, resulting in vision loss and blindness. To tackle retinal disorders, the developing retina has been explored as a versatile model to study intercellular signaling, as it presents a broad neurochemical repertoire that has been approached in the last decades in terms of signaling and diseases. Retina, dissociated and arranged as typical cultures, as mixed or neuron- and glia-enriched, and/or organized as neurospheres and/or as organoids, are valuable to understand both neuronal and glial compartments, which have contributed to revealing roles and mechanisms between transmitter systems as well as antioxidants, trophic factors, and extracellular matrix proteins. Overall, contributions in understanding neurogenesis, tissue development, differentiation, connectivity, plasticity, and cell death are widely described. A complete access to the genome of several vertebrates, as well as the recent transcriptome at the single cell level at different stages of development, also anticipates future advances in providing cues to target blinding diseases or retinal dysfunctions.
Collapse
Affiliation(s)
- Matheus H. Tempone
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Vladimir P. Borges-Martins
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Felipe César
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Dio Pablo Alexandrino-Mattos
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Camila S. de Figueiredo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Ícaro Raony
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (Í.R.); (H.R.F.)
| | - Aline Araujo dos Santos
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Aline Teixeira Duarte-Silva
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Mariana Santana Dias
- Laboratory of Gene Therapy and Viral Vectors, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.S.D.); (H.P.S.)
| | - Hércules Rezende Freitas
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (Í.R.); (H.R.F.)
| | - Elisabeth G. de Araújo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
- National Institute of Science and Technology on Neuroimmunomodulation—INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Victor Tulio Ribeiro-Resende
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| | - Marcelo Cossenza
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Hilda P. Silva
- Laboratory of Gene Therapy and Viral Vectors, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.S.D.); (H.P.S.)
| | - Roberto P. de Carvalho
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Ana L. M. Ventura
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Karin C. Calaza
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; (C.S.d.F.); (A.T.D.-S.); (E.G.d.A.); (R.P.d.C.); (A.L.M.V.); (K.C.C.)
| | - Mariana S. Silveira
- Laboratory for Investigation in Neuroregeneration and Development, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil;
| | - Regina C. C. Kubrusly
- Department of Physiology and Pharmacology, Biomedical Institute and Program of Neurosciences, Federal Fluminense University, Niterói 24020-150, Brazil; (V.P.B.-M.); (A.A.d.S.); (M.C.); (R.C.C.K.)
| | - Ricardo A. de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21949-000, Brazil; (M.H.T.); (F.C.); (D.P.A.-M.); (V.T.R.-R.)
| |
Collapse
|
22
|
Rivera-Villaseñor A, Higinio-Rodríguez F, López-Hidalgo M. Astrocytes in Pain Perception: A Systems Neuroscience Approach. ADVANCES IN NEUROBIOLOGY 2024; 39:193-212. [PMID: 39190076 DOI: 10.1007/978-3-031-64839-7_8] [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 play an active role in the function of the brain integrating neuronal activity and regulating back neuronal dynamic. They have recently emerged as active contributors of brain's emergent properties such as perceptions. Here, we analyzed the role of astrocytes in pain perception from the lens of systems neuroscience, and we do this by analyzing how astrocytes encode nociceptive information within brain processing areas and how they are key regulators of the internal state that determines pain perception. Specifically, we discuss the dynamic interactions between astrocytes and neuromodulators, such as noradrenaline, highlighting their role in shaping the level of activation of the neuronal ensemble, thereby influencing the experience of pain. Also, we will discuss the possible implications of an "Astro-NeuroMatrix" in the integration of pain across sensory, affective, and cognitive dimensions of pain perception.
Collapse
Affiliation(s)
- Angélica Rivera-Villaseñor
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico
| | - Frida Higinio-Rodríguez
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico
| | - Mónica López-Hidalgo
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico.
| |
Collapse
|
23
|
Di Palma M, Catalano M, Serpe C, De Luca M, Monaco L, Kunzelmann K, Limatola C, Conti F, Fattorini G. Lipopolysaccharide augments microglial GABA uptake by increasing GABA transporter-1 trafficking and bestrophin-1 expression. Glia 2023; 71:2527-2540. [PMID: 37431178 DOI: 10.1002/glia.24437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023]
Abstract
Gamma-aminobutyric acid (GABA), the principal inhibitory neurotransmitter in the brain, affects numerous immune cell functions. Microglia, the brain's resident innate immune cells, regulate GABA signaling through GABA receptors and express the complete GABAergic machinery for GABA synthesis, uptake, and release. Here, the use of primary microglial cell cultures and ex vivo brain tissue sections allowed for demonstrating that treatment with lipopolysaccharide (LPS) increased microglial GABA uptake as well as GABA transporter (GAT)-1 trafficking. This effect was not entirely abolished by treatment with GAT inhibitors (GAT-Is). Notably, LPS also induced microglial upregulation of bestrophin-1 (BEST-1), a Ca2+ -activated Cl- channel permeable to GABA. Combined administration of GAT-Is and a BEST-1 inhibitor completely abolished LPS-induced microglial GABA uptake. Interestingly, increased microglial GAT-1 membrane turnover via syntaxin 1A was detected in LPS-treated cultures after BEST-1 blockade. Altogether, these findings provided evidence for a novel mechanism through which LPS may trigger the inflammatory response by directly altering microglial GABA clearance and identified the GAT-1/BEST-1 interplay as a potential novel mechanism involved in brain inflammation.
Collapse
Affiliation(s)
- Michael Di Palma
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Carmela Serpe
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Mariassunta De Luca
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, Regensburg, Germany
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
- Center for Neurobiology of Aging, INRCA IRCCS, Ancona, Italy
- Fondazione di Medicina Molecolare, Università Politecnica delle Marche, Ancona, Italy
| | - Giorgia Fattorini
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
- Center for Neurobiology of Aging, INRCA IRCCS, Ancona, Italy
| |
Collapse
|
24
|
Verkhratsky A, Butt A, Li B, Illes P, Zorec R, Semyanov A, Tang Y, Sofroniew MV. Astrocytes in human central nervous system diseases: a frontier for new therapies. Signal Transduct Target Ther 2023; 8:396. [PMID: 37828019 PMCID: PMC10570367 DOI: 10.1038/s41392-023-01628-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 10/14/2023] Open
Abstract
Astroglia are a broad class of neural parenchymal cells primarily dedicated to homoeostasis and defence of the central nervous system (CNS). Astroglia contribute to the pathophysiology of all neurological and neuropsychiatric disorders in ways that can be either beneficial or detrimental to disorder outcome. Pathophysiological changes in astroglia can be primary or secondary and can result in gain or loss of functions. Astroglia respond to external, non-cell autonomous signals associated with any form of CNS pathology by undergoing complex and variable changes in their structure, molecular expression, and function. In addition, internally driven, cell autonomous changes of astroglial innate properties can lead to CNS pathologies. Astroglial pathophysiology is complex, with different pathophysiological cell states and cell phenotypes that are context-specific and vary with disorder, disorder-stage, comorbidities, age, and sex. Here, we classify astroglial pathophysiology into (i) reactive astrogliosis, (ii) astroglial atrophy with loss of function, (iii) astroglial degeneration and death, and (iv) astrocytopathies characterised by aberrant forms that drive disease. We review astroglial pathophysiology across the spectrum of human CNS diseases and disorders, including neurotrauma, stroke, neuroinfection, autoimmune attack and epilepsy, as well as neurodevelopmental, neurodegenerative, metabolic and neuropsychiatric disorders. Characterising cellular and molecular mechanisms of astroglial pathophysiology represents a new frontier to identify novel therapeutic strategies.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania.
| | - Arthur Butt
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Peter Illes
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04109, Leipzig, Germany
| | - Robert Zorec
- Celica Biomedical, Lab Cell Engineering, Technology Park, 1000, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Alexey Semyanov
- Department of Physiology, Jiaxing University College of Medicine, 314033, Jiaxing, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education/Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Lia A, Di Spiezio A, Vitalini L, Tore M, Puja G, Losi G. Ion Channels and Ionotropic Receptors in Astrocytes: Physiological Functions and Alterations in Alzheimer's Disease and Glioblastoma. Life (Basel) 2023; 13:2038. [PMID: 37895420 PMCID: PMC10608464 DOI: 10.3390/life13102038] [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: 09/04/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
The human brain is composed of nearly one hundred billion neurons and an equal number of glial cells, including macroglia, i.e., astrocytes and oligodendrocytes, and microglia, the resident immune cells of the brain. In the last few decades, compelling evidence has revealed that glial cells are far more active and complex than previously thought. In particular, astrocytes, the most abundant glial cell population, not only take part in brain development, metabolism, and defense against pathogens and insults, but they also affect sensory, motor, and cognitive functions by constantly modulating synaptic activity. Not surprisingly, astrocytes are actively involved in neurodegenerative diseases (NDs) and other neurological disorders like brain tumors, in which they rapidly become reactive and mediate neuroinflammation. Reactive astrocytes acquire or lose specific functions that differently modulate disease progression and symptoms, including cognitive impairments. Astrocytes express several types of ion channels, including K+, Na+, and Ca2+ channels, transient receptor potential channels (TRP), aquaporins, mechanoreceptors, and anion channels, whose properties and functions are only partially understood, particularly in small processes that contact synapses. In addition, astrocytes express ionotropic receptors for several neurotransmitters. Here, we provide an extensive and up-to-date review of the roles of ion channels and ionotropic receptors in astrocyte physiology and pathology. As examples of two different brain pathologies, we focus on Alzheimer's disease (AD), one of the most diffuse neurodegenerative disorders, and glioblastoma (GBM), the most common brain tumor. Understanding how ion channels and ionotropic receptors in astrocytes participate in NDs and tumors is necessary for developing new therapeutic tools for these increasingly common neurological conditions.
Collapse
Affiliation(s)
- Annamaria Lia
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
| | - Alessandro Di Spiezio
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
- Neuroscience Institute (CNR-IN), Padova Section, 35131 Padova, Italy
| | - Lorenzo Vitalini
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Manuela Tore
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Puja
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Gabriele Losi
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
26
|
Lemcke R, Egebjerg C, Berendtsen NT, Egerod KL, Thomsen AR, Pers TH, Christensen JP, Kornum BR. Molecular consequences of peripheral Influenza A infection on cell populations in the murine hypothalamus. eLife 2023; 12:RP87515. [PMID: 37698546 PMCID: PMC10497288 DOI: 10.7554/elife.87515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023] Open
Abstract
Infection with Influenza A virus (IAV) causes the well-known symptoms of the flu, including fever, loss of appetite, and excessive sleepiness. These responses, mediated by the brain, will normally disappear once the virus is cleared from the system, but a severe respiratory virus infection may cause long-lasting neurological disturbances. These include encephalitis lethargica and narcolepsy. The mechanisms behind such long lasting changes are unknown. The hypothalamus is a central regulator of the homeostatic response during a viral challenge. To gain insight into the neuronal and non-neuronal molecular changes during an IAV infection, we intranasally infected mice with an H1N1 virus and extracted the brain at different time points. Using single-nucleus RNA sequencing (snRNA-seq) of the hypothalamus, we identify transcriptional effects in all identified cell populations. The snRNA-seq data showed the most pronounced transcriptional response at 3 days past infection, with a strong downregulation of genes across all cell types. General immune processes were mainly impacted in microglia, the brain resident immune cells, where we found increased numbers of cells expressing pro-inflammatory gene networks. In addition, we found that most neuronal cell populations downregulated genes contributing to the energy homeostasis in mitochondria and protein translation in the cytosol, indicating potential reduced cellular and neuronal activity. This might be a preventive mechanism in neuronal cells to avoid intracellular viral replication and attack by phagocytosing cells. The change of microglia gene activity suggest that this is complemented by a shift in microglia activity to provide increased surveillance of their surroundings.
Collapse
Affiliation(s)
- René Lemcke
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Christine Egebjerg
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Nicolai T Berendtsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Kristoffer L Egerod
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Allan R Thomsen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Jan P Christensen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| | - Birgitte R Kornum
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
27
|
Zhang YM, Qi YB, Gao YN, Chen WG, Zhou T, Zang Y, Li J. Astrocyte metabolism and signaling pathways in the CNS. Front Neurosci 2023; 17:1217451. [PMID: 37732313 PMCID: PMC10507181 DOI: 10.3389/fnins.2023.1217451] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/18/2023] [Indexed: 09/22/2023] Open
Abstract
Astrocytes comprise half of the cells in the central nervous system and play a critical role in maintaining metabolic homeostasis. Metabolic dysfunction in astrocytes has been indicated as the primary cause of neurological diseases, such as depression, Alzheimer's disease, and epilepsy. Although the metabolic functionalities of astrocytes are well known, their relationship to neurological disorders is poorly understood. The ways in which astrocytes regulate the metabolism of glucose, amino acids, and lipids have all been implicated in neurological diseases. Metabolism in astrocytes has also exhibited a significant influence on neuron functionality and the brain's neuro-network. In this review, we focused on metabolic processes present in astrocytes, most notably the glucose metabolic pathway, the fatty acid metabolic pathway, and the amino-acid metabolic pathway. For glucose metabolism, we focused on the glycolysis pathway, pentose-phosphate pathway, and oxidative phosphorylation pathway. In fatty acid metabolism, we followed fatty acid oxidation, ketone body metabolism, and sphingolipid metabolism. For amino acid metabolism, we summarized neurotransmitter metabolism and the serine and kynurenine metabolic pathways. This review will provide an overview of functional changes in astrocyte metabolism and provide an overall perspective of current treatment and therapy for neurological disorders.
Collapse
Affiliation(s)
- Yong-mei Zhang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying-bei Qi
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ya-nan Gao
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wen-gang Chen
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ting Zhou
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jia Li
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
28
|
Ueta Y, Miyata M. Functional and structural synaptic remodeling mechanisms underlying somatotopic organization and reorganization in the thalamus. Neurosci Biobehav Rev 2023; 152:105332. [PMID: 37524138 DOI: 10.1016/j.neubiorev.2023.105332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/09/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
The somatosensory system organizes the topographic representation of body maps, termed somatotopy, at all levels of an ascending hierarchy. Postnatal maturation of somatotopy establishes optimal somatosensation, whereas deafferentation in adults reorganizes somatotopy, which underlies pathological somatosensation, such as phantom pain and complex regional pain syndrome. Here, we focus on the mouse whisker somatosensory thalamus to study how sensory experience shapes the fine topography of afferent connectivity during the critical period and what mechanisms remodel it and drive a large-scale somatotopic reorganization after peripheral nerve injury. We will review our findings that, following peripheral nerve injury in adults, lemniscal afferent synapses onto thalamic neurons are remodeled back to immature configuration, as if the critical period reopens. The remodeling process is initiated with local activation of microglia in the brainstem somatosensory nucleus downstream to injured nerves and heterosynaptically controlled by input from GABAergic and cortical neurons to thalamic neurons. These fruits of thalamic studies complement well-studied cortical mechanisms of somatotopic organization and reorganization and unveil potential intervention points in treating pathological somatosensation.
Collapse
Affiliation(s)
- Yoshifumi Ueta
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Mariko Miyata
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| |
Collapse
|
29
|
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: 47] [Impact Index Per Article: 23.5] [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
|
30
|
Sa M, Yoo ES, Koh W, Park MG, Jang HJ, Yang YR, Bhalla M, Lee JH, Lim J, Won W, Kwon J, Kwon JH, Seong Y, Kim B, An H, Lee SE, Park KD, Suh PG, Sohn JW, Lee CJ. Hypothalamic GABRA5-positive neurons control obesity via astrocytic GABA. Nat Metab 2023; 5:1506-1525. [PMID: 37653043 DOI: 10.1038/s42255-023-00877-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/25/2023] [Indexed: 09/02/2023]
Abstract
The lateral hypothalamic area (LHA) regulates food intake and energy balance. Although LHA neurons innervate adipose tissues, the identity of neurons that regulate fat is undefined. Here we show that GABRA5-positive neurons in LHA (GABRA5LHA) polysynaptically project to brown and white adipose tissues in the periphery. GABRA5LHA are a distinct subpopulation of GABAergic neurons and show decreased pacemaker firing in diet-induced obesity mouse models in males. Chemogenetic inhibition of GABRA5LHA suppresses fat thermogenesis and increases weight gain, whereas gene silencing of GABRA5 in LHA decreases weight gain. In the diet-induced obesity mouse model, GABRA5LHA are tonically inhibited by nearby reactive astrocytes releasing GABA, which is synthesized by monoamine oxidase B (Maob). Gene silencing of astrocytic Maob in LHA facilitates fat thermogenesis and reduces weight gain significantly without affecting food intake, which is recapitulated by administration of a Maob inhibitor, KDS2010. We propose that firing of GABRA5LHA suppresses fat accumulation and selective inhibition of astrocytic GABA is a molecular target for treating obesity.
Collapse
Affiliation(s)
- Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea
| | - Eun-Seon Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Mingu Gordon Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Hyun-Jun Jang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Yong Ryoul Yang
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Mridula Bhalla
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Jae-Hun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Woojin Won
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Jea Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Joon-Ho Kwon
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Yejin Seong
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Byungeun Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Heeyoung An
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ki Duk Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Pann-Ghill Suh
- Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seongbuk-gu, Seoul, Republic of Korea.
- IBS School, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
31
|
Miguel-Quesada C, Zaforas M, Herrera-Pérez S, Lines J, Fernández-López E, Alonso-Calviño E, Ardaya M, Soria FN, Araque A, Aguilar J, Rosa JM. Astrocytes adjust the dynamic range of cortical network activity to control modality-specific sensory information processing. Cell Rep 2023; 42:112950. [PMID: 37543946 DOI: 10.1016/j.celrep.2023.112950] [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: 10/19/2022] [Revised: 03/21/2023] [Accepted: 07/23/2023] [Indexed: 08/08/2023] Open
Abstract
Cortical neuron-astrocyte communication in response to peripheral sensory stimulation occurs in a topographic-, frequency-, and intensity-dependent manner. However, the contribution of this functional interaction to the processing of sensory inputs and consequent behavior remains unclear. We investigate the role of astrocytes in sensory information processing at circuit and behavioral levels by monitoring and manipulating astrocytic activity in vivo. We show that astrocytes control the dynamic range of the cortical network activity, optimizing its responsiveness to incoming sensory inputs. The astrocytic modulation of sensory processing contributes to setting the detection threshold for tactile and thermal behavior responses. The mechanism of such astrocytic control is mediated through modulation of inhibitory transmission to adjust the gain and sensitivity of responding networks. These results uncover a role for astrocytes in maintaining the cortical network activity in an optimal range to control behavior associated with specific sensory modalities.
Collapse
Affiliation(s)
- Claudia Miguel-Quesada
- Neuronal Circuits and Behaviour Group, Hospital Nacional de Parapléjicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain; Experimental Neurophysiology Group, Hospital Nacional de Parapléjicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Marta Zaforas
- Experimental Neurophysiology Group, Hospital Nacional de Parapléjicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Salvador Herrera-Pérez
- Experimental Neurophysiology Group, Hospital Nacional de Parapléjicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Justin Lines
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Elena Fernández-López
- Experimental Neurophysiology Group, Hospital Nacional de Parapléjicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Elena Alonso-Calviño
- Experimental Neurophysiology Group, Hospital Nacional de Parapléjicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Maria Ardaya
- Donostia International Physics Center (DIPC), 20018 San Sebastian, Spain; Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
| | - Federico N Soria
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Juan Aguilar
- Experimental Neurophysiology Group, Hospital Nacional de Parapléjicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain.
| | - Juliana M Rosa
- Neuronal Circuits and Behaviour Group, Hospital Nacional de Parapléjicos, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain.
| |
Collapse
|
32
|
Purushotham SS, Buskila Y. Astrocytic modulation of neuronal signalling. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1205544. [PMID: 37332623 PMCID: PMC10269688 DOI: 10.3389/fnetp.2023.1205544] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023]
Abstract
Neuronal signalling is a key element in neuronal communication and is essential for the proper functioning of the CNS. Astrocytes, the most prominent glia in the brain play a key role in modulating neuronal signalling at the molecular, synaptic, cellular, and network levels. Over the past few decades, our knowledge about astrocytes and their functioning has evolved from considering them as merely a brain glue that provides structural support to neurons, to key communication elements. Astrocytes can regulate the activity of neurons by controlling the concentrations of ions and neurotransmitters in the extracellular milieu, as well as releasing chemicals and gliotransmitters that modulate neuronal activity. The aim of this review is to summarise the main processes through which astrocytes are modulating brain function. We will systematically distinguish between direct and indirect pathways in which astrocytes affect neuronal signalling at all levels. Lastly, we will summarize pathological conditions that arise once these signalling pathways are impaired focusing on neurodegeneration.
Collapse
Affiliation(s)
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- The MARCS Institute, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
33
|
Ueberbach T, Simacek CA, Tegeder I, Kirischuk S, Mittmann T. Tonic activation of GABA B receptors via GAT-3 mediated GABA release reduces network activity in the developing somatosensory cortex in GAD67-GFP mice. Front Synaptic Neurosci 2023; 15:1198159. [PMID: 37325697 PMCID: PMC10267986 DOI: 10.3389/fnsyn.2023.1198159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/05/2023] [Indexed: 06/17/2023] Open
Abstract
The efficiency of neocortical information processing critically depends on the balance between the glutamatergic (excitatory, E) and GABAergic (inhibitory, I) synaptic transmission. A transient imbalance of the E/I-ratio during early development might lead to neuropsychiatric disorders later in life. The transgenic glutamic acid decarboxylase 67-green fluorescent protein (GAD67-GFP) mouse line (KI) was developed to selectively visualize GABAergic interneurons in the CNS. However, haplodeficiency of the GAD67 enzyme, the main GABA synthetizing enzyme in the brain, temporarily leads to a low GABA level in the developing brain of these animals. However, KI mice did not demonstrate any epileptic activity and only few and mild behavioral deficits. In the present study we investigated how the developing somatosensory cortex of KI-mice compensates the reduced GABA level to prevent brain hyperexcitability. Whole-cell patch clamp recordings from layer 2/3 pyramidal neurons at P14 and at P21 revealed a reduced frequency of miniature inhibitory postsynaptic currents (mIPSCs) in KI mice without any change in amplitude or kinetics. Interestingly, mEPSC frequencies were also decreased, while the E/I-ratio was nevertheless shifted toward excitation. Surprisingly, multi-electrode-recordings (MEA) from acute slices revealed a decreased spontaneous neuronal network activity in KI mice compared to wild-type (WT) littermates, pointing to a compensatory mechanism that prevents hyperexcitability. Blockade of GABAB receptors (GABABRs) with CGP55845 strongly increased the frequency of mEPSCs in KI, but failed to affect mIPSCs in any genotype or age. It also induced a membrane depolarization in P14 KI, but not in P21 KI or WT mice. MEA recordings in presence of CGP55845 revealed comparable levels of network activity in both genotypes, indicating that tonically activated GABABRs balance neuronal activity in P14 KI cortex despite the reduced GABA levels. Blockade of GABA transporter 3 (GAT-3) reproduced the CGP55845 effects suggesting that tonic activation of GABABRs is mediated by ambient GABA released via GAT-3 operating in reverse mode. We conclude that GAT-3-mediated GABA release leads to tonic activation of both pre- and postsynaptic GABABRs and restricts neuronal excitability in the developing cortex to compensate for reduced neuronal GABA synthesis. Since GAT-3 is predominantly located in astrocytes, GAD67 haplodeficiency may potentially stimulate astrocytic GABA synthesis through GAD67-independent pathways.
Collapse
Affiliation(s)
- Timo Ueberbach
- Institute for Physiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Clara A. Simacek
- Institute for Physiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University, Frankfurt, Germany
| | - Sergei Kirischuk
- Institute for Physiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Mittmann
- Institute for Physiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
34
|
Stasenko SV, Kazantsev VB. Information Encoding in Bursting Spiking Neural Network Modulated by Astrocytes. ENTROPY (BASEL, SWITZERLAND) 2023; 25:e25050745. [PMID: 37238500 DOI: 10.3390/e25050745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023]
Abstract
We investigated a mathematical model composed of a spiking neural network (SNN) interacting with astrocytes. We analysed how information content in the form of two-dimensional images can be represented by an SNN in the form of a spatiotemporal spiking pattern. The SNN includes excitatory and inhibitory neurons in some proportion, sustaining the excitation-inhibition balance of autonomous firing. The astrocytes accompanying each excitatory synapse provide a slow modulation of synaptic transmission strength. An information image was uploaded to the network in the form of excitatory stimulation pulses distributed in time reproducing the shape of the image. We found that astrocytic modulation prevented stimulation-induced SNN hyperexcitation and non-periodic bursting activity. Such homeostatic astrocytic regulation of neuronal activity makes it possible to restore the image supplied during stimulation and lost in the raster diagram of neuronal activity due to non-periodic neuronal firing. At a biological point, our model shows that astrocytes can act as an additional adaptive mechanism for regulating neural activity, which is crucial for sensory cortical representations.
Collapse
Affiliation(s)
- Sergey V Stasenko
- Laboratory of Advanced Methods for High-Dimensional Data Analysis, Lobachevsky State University of Nizhny Novgorod, 603022 Nizhny Novgorod, Russia
| | - Victor B Kazantsev
- Laboratory of Advanced Methods for High-Dimensional Data Analysis, Lobachevsky State University of Nizhny Novgorod, 603022 Nizhny Novgorod, Russia
| |
Collapse
|
35
|
Jung H, Lee D, You H, Lee M, Kim H, Cheong E, Um JW. LPS induces microglial activation and GABAergic synaptic deficits in the hippocampus accompanied by prolonged cognitive impairment. Sci Rep 2023; 13:6547. [PMID: 37085584 PMCID: PMC10121592 DOI: 10.1038/s41598-023-32798-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/03/2023] [Indexed: 04/23/2023] Open
Abstract
Neuroinflammation impacts the brain and cognitive behavior through microglial activation. In this study, we determined the temporal sequence from microglial activation to synaptic dysfunction and cognitive behavior induced by neuroinflammation in mice. We found that LPS injection activated microglia within a short period, followed by impairments in GABAergic synapses, and that these events led to long-term cognitive impairment. We demonstrated that, 3 days after LPS injection, microglia in the hippocampus were significantly activated due to the LPS-induced inflammation in association with alterations in cellular morphology, microglial density, and expression of phagocytic markers. GABAergic synaptic impairments were detected at 4-6 days after LPS treatment, a time when microglia activity had returned to normal. Consequently, memory impairment persisted for 6 days after injection of LPS. Our results suggest that neuroinflammation induces microglia activation, GABAergic synaptic deficits and prolonged memory impairment over a defined temporal sequence. Our observations provide insight into the temporal sequence of neuroinflammation-associated brain pathologies. Moreover, the specific loss of inhibitory synapses accompanying the impaired inhibitory synaptic transmission provides mechanistic insight that may explain the prolonged cognitive deficit observed in patients with neuroinflammation. Thus, this study provides essential clues regarding early intervention strategies against brain pathologies accompanying neuroinflammation.
Collapse
Affiliation(s)
- Hyeji Jung
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea
| | - Dongsu Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Heejung You
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Myungha Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Hyeonho Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea.
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea.
- Center for Synapse Diversity and Specificity, DGIST, 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu, 42988, Korea.
| |
Collapse
|
36
|
Cheng YT, Woo J, Luna-Figueroa E, Maleki E, Harmanci AS, Deneen B. Social deprivation induces astrocytic TRPA1-GABA suppression of hippocampal circuits. Neuron 2023; 111:1301-1315.e5. [PMID: 36787749 PMCID: PMC10121837 DOI: 10.1016/j.neuron.2023.01.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 12/13/2022] [Accepted: 01/18/2023] [Indexed: 02/16/2023]
Abstract
Social experience is essential for the development and maintenance of higher-order brain function. Social deprivation results in a host of cognitive deficits, and cellular studies have largely focused on associated neuronal dysregulation; how astrocyte function is impacted by social deprivation is unknown. Here, we show that hippocampal astrocytes from juvenile mice subjected to social isolation exhibit increased Ca2+ activity and global changes in gene expression. We found that the Ca2+ channel TRPA1 is upregulated in astrocytes after social deprivation and astrocyte-specific deletion of TRPA1 reverses the physiological and cognitive deficits associated with social deprivation. Mechanistically, TRPA1 inhibition of hippocampal circuits is mediated by a parallel increase of astrocytic production and release of the inhibitory neurotransmitter GABA after social deprivation. Collectively, our studies reveal how astrocyte function is tuned to social experience and identifies a social-context-specific mechanism by which astrocytic TRPA1 and GABA coordinately suppress hippocampal circuit function.
Collapse
Affiliation(s)
- Yi-Ting Cheng
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Junsung Woo
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Estefania Luna-Figueroa
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ehson Maleki
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Benjamin Deneen
- Center for Cancer Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
37
|
Yang J, Chen J, Liu Y, Chen KH, Baraban JM, Qiu Z. Ventral tegmental area astrocytes modulate cocaine reward by tonically releasing GABA. Neuron 2023; 111:1104-1117.e6. [PMID: 36681074 PMCID: PMC10079641 DOI: 10.1016/j.neuron.2022.12.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/30/2022] [Accepted: 12/27/2022] [Indexed: 01/21/2023]
Abstract
Addictive drugs increase ventral tegmental area (VTA) dopamine (DA) neuron activity through distinct cellular mechanisms, one of which involves disinhibition of DA neurons by inhibiting local GABA neurons. How drugs regulate VTA GABA neuron activity and drive addictive behaviors remains poorly understood. Here, we show that astrocytes control VTA GABA neuron activity in cocaine reward via tonic inhibition in mice. Repeated cocaine exposure potentiates astrocytic tonic GABA release through volume-regulated anion channels (VRACs) and augments tonic inhibition of VTA GABA neurons, thus downregulating their activities and disinhibiting nucleus accumbens (NAc) projecting DA neurons. Attenuation of tonic inhibition by either deleting Swell1 (Lrrc8a), the obligatory subunit of VRACs, in VTA astrocytes or disrupting δ subunit of GABAA receptors in VTA GABA neurons reduces cocaine-evoked changes in neuron activity, locomotion, and reward behaviors in mice. Together, our findings reveal the critical role of astrocytes in regulating the VTA local circuit and cocaine reward.
Collapse
Affiliation(s)
- Junhua Yang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jianan Chen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yongqing Liu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kevin Hong Chen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jay M Baraban
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhaozhu Qiu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
38
|
Tang Y, Yan Y, Mao J, Ni J, Qing H. The hippocampus associated GABAergic neural network impairment in early-stage of Alzheimer's disease. Ageing Res Rev 2023; 86:101865. [PMID: 36716975 DOI: 10.1016/j.arr.2023.101865] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is the commonest neurodegenerative disease with slow progression. Pieces of evidence suggest that the GABAergic system is impaired in the early stage of AD, leading to hippocampal neuron over-activity and further leading to memory and cognitive impairment in patients with AD. However, the precise impairment mechanism of the GABAergic system on the pathogenesis of AD is still unclear. The impairment of neural networks associated with the GABAergic system is tightly associated with AD. Therefore, we describe the roles played by hippocampus-related GABAergic circuits and their impairments in AD neuropathology. In addition, we give our understand on the process from GABAergic circuit impairment to cognitive and memory impairment, since recent studies on astrocyte in AD plays an important role behind cognition dysfunction caused by GABAergic circuit impairment, which helps better understand the GABAergic system and could open up innovative AD therapy.
Collapse
Affiliation(s)
- Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jian Mao
- Zhengzhou Tobacco Institute of China National Tobacco Company, Zhengzhou 450001, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Department of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China.
| |
Collapse
|
39
|
Lia A, Sansevero G, Chiavegato A, Sbrissa M, Pendin D, Mariotti L, Pozzan T, Berardi N, Carmignoto G, Fasolato C, Zonta M. Rescue of astrocyte activity by the calcium sensor STIM1 restores long-term synaptic plasticity in female mice modelling Alzheimer's disease. Nat Commun 2023; 14:1590. [PMID: 36949142 PMCID: PMC10033875 DOI: 10.1038/s41467-023-37240-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 03/09/2023] [Indexed: 03/24/2023] Open
Abstract
Calcium dynamics in astrocytes represent a fundamental signal that through gliotransmitter release regulates synaptic plasticity and behaviour. Here we present a longitudinal study in the PS2APP mouse model of Alzheimer's disease (AD) linking astrocyte Ca2+ hypoactivity to memory loss. At the onset of plaque deposition, somatosensory cortical astrocytes of AD female mice exhibit a drastic reduction of Ca2+ signaling, closely associated with decreased endoplasmic reticulum Ca2+ concentration and reduced expression of the Ca2+ sensor STIM1. In parallel, astrocyte-dependent long-term synaptic plasticity declines in the somatosensory circuitry, anticipating specific tactile memory loss. Notably, we show that both astrocyte Ca2+ signaling and long-term synaptic plasticity are fully recovered by selective STIM1 overexpression in astrocytes. Our data unveil astrocyte Ca2+ hypoactivity in neocortical astrocytes as a functional hallmark of early AD stages and indicate astrocytic STIM1 as a target to rescue memory deficits.
Collapse
Affiliation(s)
- Annamaria Lia
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Gabriele Sansevero
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Angela Chiavegato
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Miriana Sbrissa
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Diana Pendin
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Letizia Mariotti
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Tullio Pozzan
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, Padua, Italy
| | - Nicoletta Berardi
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Giorgio Carmignoto
- Neuroscience Institute, National Research Council (CNR), Padua, Italy.
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| | - Micaela Zonta
- Neuroscience Institute, National Research Council (CNR), Padua, Italy.
- Department of Biomedical Sciences, University of Padua, Padua, Italy.
| |
Collapse
|
40
|
Astrocytes regulate inhibitory neurotransmission through GABA uptake, metabolism, and recycling. Essays Biochem 2023; 67:77-91. [PMID: 36806927 DOI: 10.1042/ebc20220208] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 02/23/2023]
Abstract
Synaptic regulation of the primary inhibitory neurotransmitter γ-aminobutyric acid (GABA) is essential for brain function. Cerebral GABA homeostasis is tightly regulated through multiple mechanisms and is directly coupled to the metabolic collaboration between neurons and astrocytes. In this essay, we outline and discuss the fundamental roles of astrocytes in regulating synaptic GABA signaling. A major fraction of synaptic GABA is removed from the synapse by astrocytic uptake. Astrocytes utilize GABA as a metabolic substrate to support glutamine synthesis. The astrocyte-derived glutamine is subsequently transferred to neurons where it serves as the primary precursor of neuronal GABA synthesis. The flow of GABA and glutamine between neurons and astrocytes is collectively termed the GABA-glutamine cycle and is essential to sustain GABA synthesis and inhibitory signaling. In certain brain areas, astrocytes are even capable of synthesizing and releasing GABA to modulate inhibitory transmission. The majority of oxidative GABA metabolism in the brain takes place in astrocytes, which also leads to synthesis of the GABA-related metabolite γ-hydroxybutyric acid (GHB). The physiological roles of endogenous GHB remain unclear, but may be related to regulation of tonic inhibition and synaptic plasticity. Disrupted inhibitory signaling and dysfunctional astrocyte GABA handling are implicated in several diseases including epilepsy and Alzheimer's disease. Synaptic GABA homeostasis is under astrocytic control and astrocyte GABA uptake, metabolism, and recycling may therefore serve as relevant targets to ameliorate pathological inhibitory signaling.
Collapse
|
41
|
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: 24] [Impact Index Per Article: 12.0] [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
|
42
|
Peng W, Liu X, Ma G, Wu Z, Wang Z, Fei X, Qin M, Wang L, Li Y, Zhang S, Xu M. Adenosine-independent regulation of the sleep-wake cycle by astrocyte activity. Cell Discov 2023; 9:16. [PMID: 36746933 PMCID: PMC9902472 DOI: 10.1038/s41421-022-00498-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 11/20/2022] [Indexed: 02/08/2023] Open
Abstract
Astrocytes play a crucial role in regulating sleep-wake behavior, and adenosine signaling is generally thought to be involved. Here we show multiple lines of evidence supporting that modulation of the sleep-wake behavior by astrocyte Ca2+ activity could occur without adenosine signaling. In the basal forebrain and the brainstem, two brain regions that are known to be essential for sleep-wake regulation, chemogenetically-induced astrocyte Ca2+ elevation significantly modulated the sleep-wake cycle. Although astrocyte Ca2+ level positively correlated with the amount of extracellular adenosine, as revealed by a genetically encoded adenosine sensor, we found no detectable change in adenosine level after suppressing astrocyte Ca2+ elevation, and transgenic mice lacking one of the major extracellular ATP-adenosine conversion enzymes showed similar extracellular adenosine level and astrocyte Ca2+-induced sleep modulation. Furthermore, astrocyte Ca2+ is dependent primarily on local neuronal activity, causing brain region-specific regulation of the sleep-wake cycle. Thus, neural activity-dependent astrocyte activity could regulate the sleep-wake behavior independent of adenosine signaling.
Collapse
Affiliation(s)
- Wanling Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China.
| | - Xiaotong Liu
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Guofen Ma
- grid.16821.3c0000 0004 0368 8293Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaofa Wu
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ziyue Wang
- grid.16821.3c0000 0004 0368 8293Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Fei
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, China
| | - Meiling Qin
- grid.9227.e0000000119573309Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Lizhao Wang
- grid.16821.3c0000 0004 0368 8293Center for Brain Science of Shanghai Children’s Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yulong Li
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Siyu Zhang
- Center for Brain Science of Shanghai Children's Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Min Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shangha, China.
| |
Collapse
|
43
|
Fletcher PA, Smiljanic K, Prévide RM, Constantin S, Sherman AS, Coon SL, Stojilkovic SS. The astroglial and stem cell functions of adult rat folliculostellate cells. Glia 2023; 71:205-228. [PMID: 36093576 PMCID: PMC9772113 DOI: 10.1002/glia.24267] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 02/06/2023]
Abstract
The mammalian pituitary gland is a complex organ consisting of hormone-producing cells, anterior lobe folliculostellate cells (FSCs), posterior lobe pituicytes, vascular pericytes and endothelial cells, and Sox2-expressing stem cells. We present single-cell RNA sequencing and immunohistofluorescence analyses of pituitary cells of adult female rats with a focus on the transcriptomic profiles of nonhormonal cell types. Samples obtained from whole pituitaries and separated anterior and posterior lobe cells contained all expected pituitary resident cell types and lobe-specific vascular cell subpopulations. FSCs and pituicytes expressed S100B, ALDOC, EAAT1, ALDH1A1, and VIM genes and proteins, as well as other astroglial marker genes, some common and some cell type-specific. We also found that the SOX2 gene and protein were expressed in ~15% of pituitary cells, including FSCs, pituicytes, and a fraction of hormone-producing cells, arguing against its stem cell specificity. FSCs comprised two Sox2-expressing subclusters; FS1 contained more cells but lower genetic diversity, while FS2 contained proliferative cells, shared genes with hormone-producing cells, and expressed genes consistent with stem cell niche formation, regulation of cell proliferation and stem cell pluripotency, including the Hippo and Wnt pathways. FS1 cells were randomly distributed in the anterior and intermediate lobes, while FS2 cells were localized exclusively in the marginal zone between the anterior and intermediate lobes. These data indicate the identity of the FSCs as anterior pituitary-specific astroglia, with FS1 cells representing differentiated cells equipped for classical FSC roles and FS2 cells exhibiting additional stem cell-like features.
Collapse
Affiliation(s)
- Patrick A. Fletcher
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Kosara Smiljanic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Rafael M. Prévide
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Stephanie Constantin
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Arthur S. Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD 20892
| | - Steven L. Coon
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, 20892
| | - Stanko S. Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD 20892
| |
Collapse
|
44
|
Kruyer A, Kalivas PW, Scofield MD. Astrocyte regulation of synaptic signaling in psychiatric disorders. Neuropsychopharmacology 2023; 48:21-36. [PMID: 35577914 PMCID: PMC9700696 DOI: 10.1038/s41386-022-01338-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 02/07/2023]
Abstract
Over the last 15 years, the field of neuroscience has evolved toward recognizing the critical role of astroglia in shaping neuronal synaptic activity and along with the pre- and postsynapse is now considered an equal partner in tripartite synaptic transmission and plasticity. The relative youth of this recognition and a corresponding deficit in reagents and technologies for quantifying and manipulating astroglia relative to neurons continues to hamper advances in understanding tripartite synaptic physiology. Nonetheless, substantial advances have been made and are reviewed herein. We review the role of astroglia in synaptic function and regulation of behavior with an eye on how tripartite synapses figure into brain pathologies underlying behavioral impairments in psychiatric disorders, both from the perspective of measures in postmortem human brains and more subtle influences on tripartite synaptic regulation of behavior in animal models of psychiatric symptoms. Our goal is to provide the reader a well-referenced state-of-the-art understanding of current knowledge and predict what we may discover with deeper investigation of tripartite synapses using reagents and technologies not yet available.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
45
|
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: 26] [Impact Index Per Article: 13.0] [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
|
46
|
Kilb W, Kirischuk S. GABA Release from Astrocytes in Health and Disease. Int J Mol Sci 2022; 23:ijms232415859. [PMID: 36555501 PMCID: PMC9784789 DOI: 10.3390/ijms232415859] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Astrocytes are the most abundant glial cells in the central nervous system (CNS) mediating a variety of homeostatic functions, such as spatial K+ buffering or neurotransmitter reuptake. In addition, astrocytes are capable of releasing several biologically active substances, including glutamate and GABA. Astrocyte-mediated GABA release has been a matter of debate because the expression level of the main GABA synthesizing enzyme glutamate decarboxylase is quite low in astrocytes, suggesting that low intracellular GABA concentration ([GABA]i) might be insufficient to support a non-vesicular GABA release. However, recent studies demonstrated that, at least in some regions of the CNS, [GABA]i in astrocytes might reach several millimoles both under physiological and especially pathophysiological conditions, thereby enabling GABA release from astrocytes via GABA-permeable anion channels and/or via GABA transporters operating in reverse mode. In this review, we summarize experimental data supporting both forms of GABA release from astrocytes in health and disease, paying special attention to possible feedback mechanisms that might govern the fine-tuning of astrocytic GABA release and, in turn, the tonic GABAA receptor-mediated inhibition in the CNS.
Collapse
|
47
|
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
|
48
|
Higinio-Rodríguez F, Rivera-Villaseñor A, Calero-Vargas I, López-Hidalgo M. From nociception to pain perception, possible implications of astrocytes. Front Cell Neurosci 2022; 16:972827. [PMID: 36159392 PMCID: PMC9492445 DOI: 10.3389/fncel.2022.972827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/15/2022] [Indexed: 11/15/2022] Open
Abstract
Astrocytes are determinants for the functioning of the CNS. They respond to neuronal activity with calcium increases and can in turn modulate synaptic transmission, brain plasticity as well as cognitive processes. Astrocytes display sensory-evoked calcium responses in different brain structures related to the discriminative system of most sensory modalities. In particular, noxious stimulation evoked calcium responses in astrocytes in the spinal cord, the hippocampus, and the somatosensory cortex. However, it is not clear if astrocytes are involved in pain. Pain is a private, personal, and complex experience that warns us about potential tissue damage. It is a perception that is not linearly associated with the amount of tissue damage or nociception; instead, it is constructed with sensory, cognitive, and affective components and depends on our previous experiences. However, it is not fully understood how pain is created from nociception. In this perspective article, we provide an overview of the mechanisms and neuronal networks that underlie the perception of pain. Then we proposed that coherent activity of astrocytes in the spinal cord and pain-related brain areas could be important in binding sensory, affective, and cognitive information on a slower time scale.
Collapse
Affiliation(s)
- Frida Higinio-Rodríguez
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Querétaro, Mexico
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Angélica Rivera-Villaseñor
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Querétaro, Mexico
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Isnarhazni Calero-Vargas
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Querétaro, Mexico
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Mónica López-Hidalgo
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Querétaro, Mexico
- *Correspondence: Mónica López-Hidalgo,
| |
Collapse
|
49
|
Choi J, Kim YS, Kim MH, Kim HJ, Yoon BE. Maternal lead exposure induces sex-dependent cerebellar glial alterations and repetitive behaviors. Front Cell Neurosci 2022; 16:954807. [PMID: 36072563 PMCID: PMC9442054 DOI: 10.3389/fncel.2022.954807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Lead (Pb) is one of the most prevalent heavy metals we encounter daily. Although there are many reports regarding their toxic effects on humans, the effects of exposure to low lead concentrations throughout the pregnancy period on the offspring are not fully elucidated yet. This study aimed to investigate the cellular mechanisms that occur in response to lead exposure. To this end, we administered lead-containing water to pregnant mice from the day of conception till delivery or till day 28 postnatally. Furthermore, we performed neurodevelopmental disorder-related behavior tests and RNA-sequencing analysis. We used both genders for all experiments because neurodevelopmental disorders usually show several sex-dependent differences. The results revealed increased levels of gliosis in the cerebella of lead-exposed pups compared to those in littermates belonging to the control group. Additionally, we observed altered behaviors of male mice in the autism spectrum disorder-related tests. RNA-sequencing results revealed changes in gamma-aminobutyric acid (GABA) signaling in the lead-exposed mouse model. Specifically, the lead-exposed male mice showed decreased monoamine oxidase B and increased levels of diamine oxidase enzyme, which is related to the synthesis of GABA in astrocytes. These findings demonstrate sex-dependent basal developmental changes in glial cells and an increased prevalence of autistic-like behaviors in the young pups of mothers exposed to lead during pregnancy.
Collapse
Affiliation(s)
- Juwon Choi
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan, South Korea
| | - Yoo Sung Kim
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan, South Korea
| | - Mi-Hye Kim
- Department of Physiology, College of Medicine, Dankook University, Cheonan, South Korea
- Department of Medical Laser, Graduate School, Dankook University, Cheonan, South Korea
| | - Hee Jung Kim
- Department of Physiology, College of Medicine, Dankook University, Cheonan, South Korea
| | - Bo-Eun Yoon
- Department of Molecular Biology, College of Natural Sciences, Dankook University, Cheonan, South Korea
- *Correspondence: Bo-Eun Yoon,
| |
Collapse
|
50
|
Inhibiting peripheral and central MAO-B ameliorates joint inflammation and cognitive impairment in rheumatoid arthritis. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1188-1200. [PMID: 35982301 PMCID: PMC9440195 DOI: 10.1038/s12276-022-00830-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/07/2022] [Accepted: 05/31/2022] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder characterized by chronic inflammation and the destruction of joints and systemic organs. RA is commonly accompanied by neuropsychiatric complications, such as cognitive impairment and depression. However, the role of monoamine oxidase (MAO) and its inhibitors in controlling neurotransmitters associated with these complications in RA have not been clearly identified. Here, we report that peripheral and central MAO-B are highly associated with joint inflammation and cognitive impairment in RA, respectively. Ribonucleic acid (RNA) sequencing and protein expression quantification were used to show that MAO-B and related molecules, such as gamma aminobutyric acid (GABA), were elevated in the inflamed synovium of RA patients. In primary cultured fibroblast-like synoviocytes in the RA synovium, MAO-B expression was significantly increased by tumor necrosis factor (TNF)-α-induced autophagy, which produces putrescine, the polyamine substrate for GABA synthesis. We also observed that MAO-B-mediated aberrant astrocytic production of GABA was augmented by interleukin (IL)-1β and inhibited CA1-hippocampal pyramidal neurons, which are responsible for memory storage, in an animal model of RA. Moreover, a newly developed reversible inhibitor of MAO-B ameliorated joint inflammation by inhibiting cyclooxygenase (Cox)-2. Therefore, MAO-B can be an effective therapeutic target for joint inflammation and cognitive impairment in patients with RA. Inhibiting an enzyme that is upregulated during joint inflammation may prove a valuable therapy for rheumatoid arthritis (RA). As well as causing considerable pain and discomfort in the joints, RA can also trigger neuropsychiatric problems including depression and memory impairment. The monoamine oxidase (MAO) enzyme family is involved in the control of neurotransmitters, and there is evidence that links MAO-B levels with systemic inflammation. C. Justin Lee at Center for Cognition and Sociality, Institute for Basic Science,, Daejeon, South Korea, and co-workers examined the role of MAO-B in RA using patient tissue samples and mouse models. MAO-B and related molecules were upregulated in patients’ inflamed joint tissues. In mice, elevated MAO-B triggered the inhibition of nerve cell activity related to memory storage. A novel drug that inhibits MAO-B reduced RA-related inflammation and cognitive impairment in mice, suggesting a promising approach to treatment.
Collapse
|