1
|
Arrigo A, Cremona O, Aragona E, Casoni F, Consalez G, Dogru RM, Hauck SM, Antropoli A, Bianco L, Parodi MB, Bandello F, Grosche A. Müller cells trophism and pathology as the next therapeutic targets for retinal diseases. Prog Retin Eye Res 2025; 106:101357. [PMID: 40254246 DOI: 10.1016/j.preteyeres.2025.101357] [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: 02/20/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Müller cells are a crucial retinal cell type involved in multiple regulatory processes and functions that are essential for retinal health and functionality. Acting as structural and functional support for retinal neurons and photoreceptors, Müller cells produce growth factors, regulate ion and fluid homeostasis, and facilitate neuronal signaling. They play a pivotal role in retinal morphogenesis and cell differentiation, significantly contributing to macular development. Due to their radial morphology and unique cytoskeletal organization, Müller cells act as optical fibers, efficiently channeling photons directly to the photoreceptors. In response to retinal damage, Müller cells undergo specific gene expression and functional changes that serve as a first line of defense for neurons, but can also lead to unwarranted cell dysfunction, contributing to cell death and neurodegeneration. In some species, Müller cells can reactivate their developmental program, promoting retinal regeneration and plasticity-a remarkable ability that holds promising therapeutic potential if harnessed in mammals. The crucial and multifaceted roles of Müller cells-that we propose to collectively call "Müller cells trophism"-highlight the necessity of maintaining their functionality. Dysfunction of Müller cells, termed "Müller cells pathology," has been associated with a plethora of retinal diseases, including age-related macular degeneration, diabetic retinopathy, vitreomacular disorders, macular telangiectasia, and inherited retinal dystrophies. In this review, we outline how even subtle disruptions in Müller cells trophism can drive the pathological cascade of Müller cells pathology, emphasizing the need for targeted therapies to preserve retinal health and prevent disease progression.
Collapse
Affiliation(s)
- Alessandro Arrigo
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Ottavio Cremona
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Emanuela Aragona
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Filippo Casoni
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giacomo Consalez
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rüya Merve Dogru
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, 80939, Germany
| | - Alessio Antropoli
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Bianco
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesco Bandello
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| |
Collapse
|
2
|
Gonçalves-Ribeiro J, Vaz SH. The IP3R2 Knockout Mice in Behavior: A Blessing or a Curse? J Neurochem 2025; 169:e70062. [PMID: 40172184 DOI: 10.1111/jnc.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
The inositol 1,4,5-triphosphate receptor type 2 (IP3R2) plays a critical role in intracellular calcium (Ca2+) signaling, particularly in astrocytes, where it mediates Ca2+ release from the endoplasmic reticulum. This mechanism is vital for astrocytic modulation of neuronal networks, impacting synaptic transmission and broader neural circuit functions. The IP3R2 knockout (IP3R2KO) mouse model has been instrumental in unraveling the nuances of astrocytic somatic Ca2+ dynamics and their implications for brain function. Despite early findings suggesting no significant behavioral or synaptic transmission changes in IP3R2KO mice, further research highlights the model's benefit in exploring cognitive, emotional, and neurodevelopmental processes. IP3R2KO mice revealed key insights into astrocytic Ca2+ signaling diversity, encompassing bulk somatic events and localized microdomain responses, which exhibit temporal and spatial variability. These animals retain alternative Ca2+ mechanisms, likely explaining the absence of severe phenotypes in some contexts. Nevertheless, IP3R2KO mice exhibit impairments in long-term memory retention, working memory, and fear memory, alongside age-related preservation of spatial memory, linking astrocytic IP3R2 signaling to higher-order cognitive functions. Additionally, studies suggest a connection between IP3R2 pathways and depression-like behaviors, with alterations in Brain-Derived Neurotrophic Factor (BDNF) levels and GABAergic signaling, highlighting its relevance to psychiatric conditions. Despite its limitations, such as residual astrocytic Ca2+ activity and inconsistent findings, the IP3R2KO model remains a valuable tool for studying astrocytic contributions to synaptic plasticity and brain function. This underscores the importance of integrating, rather than dismissing, the IP3R2KO model in the development of new methodologies for studying astrocytic Ca2+ dynamics. The use of this model will continue to elucidate the complex interplay between astrocytes and neuronal circuits, fostering advances in understanding astrocytic Ca2+ signaling's role in health and disease.
Collapse
Affiliation(s)
- Joana Gonçalves-Ribeiro
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Centro Cardiovascular da Universidade de Lisboa, CCUL (CCUL@RISE), Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Gulbenkian Institute for Molecular Medicine (GIMM), Lisbon, Portugal
| | - Sandra H Vaz
- Faculdade de Medicina, Instituto de Farmacologia e Neurociências, Universidade de Lisboa, Lisboa, Portugal
- Centro Cardiovascular da Universidade de Lisboa, CCUL (CCUL@RISE), Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Gulbenkian Institute for Molecular Medicine (GIMM), Lisbon, Portugal
| |
Collapse
|
3
|
Wu M, Zhang R, Fu P, Mei Y. Disrupted astrocyte-neuron signaling reshapes brain activity in epilepsy and Alzheimer's disease. Neuroscience 2025; 570:132-151. [PMID: 39986432 DOI: 10.1016/j.neuroscience.2025.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/20/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Astrocytes establish dynamic interactions with surrounding neurons and synchronize neuronal networks within a specific range. However, these reciprocal astrocyte-neuronal interactions are selectively disrupted in epilepsy and Alzheimer's disease (AD), which contributes to the initiation and progression of network hypersynchrony. Deciphering how disrupted astrocyte-neuronal signaling reshapes brain activity is crucial to prevent subclinical epileptiform activity in epilepsy and AD. In this review, we provide an overview of the diverse astrocyte-neuronal crosstalk in maintaining of network activity via homeostatic control of extracellular ions and transmitters, synapse formation and elimination. More importantly, since AD and epilepsy share the common symptoms of neuronal hyperexcitability and astrogliosis, we then explore the crosstalk between astrocytes and neurons in the context of epilepsy and AD and discuss how these disrupted interactions reshape brain activity in pathological conditions. Collectively, this review sheds light on how disrupted astrocyte-neuronal signaling reshapes brain activity in epilepsy and AD, and highlights that modifying astrocyte-neuronal signaling could be a therapeutic approach to prevent epileptiform activity in AD.
Collapse
Affiliation(s)
- Mengjie Wu
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ruonan Zhang
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Peng Fu
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
4
|
Gobbo D, Kirchhoff F. Animal-based approaches to understanding neuroglia physiology in vitro and in vivo. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:229-263. [PMID: 40122627 DOI: 10.1016/b978-0-443-19104-6.00012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter describes the pivotal role of animal models for unraveling the physiology of neuroglial cells in the central nervous system (CNS). The two rodent species Mus musculus (mice) and Rattus norvegicus (rats) have been indispensable in scientific research due to their remarkable resemblance to humans anatomically, physiologically, and genetically. Their ease of maintenance, short gestation times, and rapid development make them ideal candidates for studying the physiology of astrocytes, oligodendrocyte-lineage cells, and microglia. Moreover, their genetic similarity to humans facilitates the investigation of molecular mechanisms governing neural physiology. Mice are largely the predominant model of neuroglial research, owing to advanced genetic manipulation techniques, whereas rats remain invaluable for applications requiring larger CNS structures for surgical manipulations. Next to rodents, other animal models, namely, Danio rerio (zebrafish) and Drosophila melanogaster (fruit fly), will be discussed to emphasize their critical role in advancing our understanding of glial physiology. Each animal model provides distinct advantages and disadvantages. By combining the strengths of each of them, researchers can gain comprehensive insights into glial function across species, ultimately promoting the understanding of glial physiology in the human CNS and driving the development of novel therapeutic interventions for CNS disorders.
Collapse
Affiliation(s)
- Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany.
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany; Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany.
| |
Collapse
|
5
|
Pham C, Komaki Y, Deàs-Just A, Le Gac B, Mouffle C, Franco C, Chaperon A, Vialou V, Tsurugizawa T, Cauli B, Li D. Astrocyte aquaporin mediates a tonic water efflux maintaining brain homeostasis. eLife 2024; 13:RP95873. [PMID: 39508543 PMCID: PMC11542920 DOI: 10.7554/elife.95873] [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: 11/15/2024] Open
Abstract
Brain water homeostasis not only provides a physical protection, but also determines the diffusion of chemical molecules key for information processing and metabolic stability. As a major type of glia in brain parenchyma, astrocytes are the dominant cell type expressing aquaporin water channel. How astrocyte aquaporin contributes to brain water homeostasis in basal physiology remains to be understood. We report that astrocyte aquaporin 4 (AQP4) mediates a tonic water efflux in basal conditions. Acute inhibition of astrocyte AQP4 leads to intracellular water accumulation as optically resolved by fluorescence-translated imaging in acute brain slices, and in vivo by fiber photometry in mobile mice. We then show that aquaporin-mediated constant water efflux maintains astrocyte volume and osmotic equilibrium, astrocyte and neuron Ca2+ signaling, and extracellular space remodeling during optogenetically induced cortical spreading depression. Using diffusion-weighted magnetic resonance imaging (DW-MRI), we observed that in vivo inhibition of AQP4 water efflux heterogeneously disturbs brain water homeostasis in a region-dependent manner. Our data suggest that astrocyte aquaporin, though bidirectional in nature, mediates a tonic water outflow to sustain cellular and environmental equilibrium in brain parenchyma.
Collapse
Affiliation(s)
- Cuong Pham
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris SeineParisFrance
| | - Yuji Komaki
- Central Institute for Experimental Medicine and Life ScienceKawasakiJapan
| | - Anna Deàs-Just
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris SeineParisFrance
| | - Benjamin Le Gac
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris SeineParisFrance
| | - Christine Mouffle
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris SeineParisFrance
| | - Clara Franco
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris SeineParisFrance
| | - Agnès Chaperon
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris SeineParisFrance
| | - Vincent Vialou
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris SeineParisFrance
| | - Tomokazu Tsurugizawa
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
- Faculty of Engineering, University of TsukubaTsukubaJapan
| | - Bruno Cauli
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris SeineParisFrance
| | - Dongdong Li
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris SeineParisFrance
| |
Collapse
|
6
|
Feng L, Gao L. The role of neurovascular coupling dysfunction in cognitive decline of diabetes patients. Front Neurosci 2024; 18:1375908. [PMID: 38576869 PMCID: PMC10991808 DOI: 10.3389/fnins.2024.1375908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Neurovascular coupling (NVC) is an important mechanism to ensure adequate blood supply to active neurons in the brain. NVC damage can lead to chronic impairment of neuronal function. Diabetes is characterized by high blood sugar and is considered an important risk factor for cognitive impairment. In this review, we provide fMRI evidence of NVC damage in diabetic patients with cognitive decline. Combined with the exploration of the major mechanisms and signaling pathways of NVC, we discuss the effects of chronic hyperglycemia on the cellular structure of NVC signaling, including key receptors, ion channels, and intercellular connections. Studying these diabetes-related changes in cell structure will help us understand the underlying causes behind diabetes-induced NVC damage and early cognitive decline, ultimately helping to identify the most effective drug targets for treatment.
Collapse
Affiliation(s)
| | - Ling Gao
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Park EH, Kao HY, Jourdi H, van Dijk MT, Carrillo-Segura S, Tunnell KW, Gutierrez J, Wallace EJ, Troy-Regier M, Radwan B, Lesburguères E, Alarcon JM, Fenton AA. Phencyclidine Disrupts Neural Coordination and Cognitive Control by Dysregulating Translation. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:252-263. [PMID: 38298788 PMCID: PMC10829677 DOI: 10.1016/j.bpsgos.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 02/02/2024] Open
Abstract
Background Phencyclidine (PCP) causes psychosis, is abused with increasing frequency, and was extensively used in antipsychotic drug discovery. PCP discoordinates hippocampal ensemble action potential discharge and impairs cognitive control in rats, but how this uncompetitive NMDA receptor (NMDAR) antagonist impairs cognition remains unknown. Methods The effects of PCP were investigated on hippocampal CA1 ensemble action potential discharge in vivo in urethane-anesthetized rats and during awake behavior in mice, on synaptic responses in ex vivo mouse hippocampus slices, in mice on a hippocampus-dependent active place avoidance task that requires cognitive control, and on activating the molecular machinery of translation in acute hippocampus slices. Mechanistic causality was assessed by comparing the PCP effects with the effects of inhibitors of protein synthesis, group I metabotropic glutamate receptors (mGluR1/5), and subunit-selective NMDARs. Results Consistent with ionotropic actions, PCP discoordinated CA1 ensemble action potential discharge. PCP caused hyperactivity and impaired active place avoidance, despite the rodents having learned the task before PCP administration. Consistent with metabotropic actions, PCP exaggerated protein synthesis-dependent DHPG-induced mGluR1/5-stimulated long-term synaptic depression. Pretreatment with anisomycin or the mGluR1/5 antagonist MPEP, both of which repress translation, prevented PCP-induced discoordination and the cognitive and sensorimotor impairments. PCP as well as the NR2A-containing NMDAR antagonist NVP-AAM077 unbalanced translation that engages the Akt, mTOR (mechanistic target of rapamycin), and 4EBP1 translation machinery and increased protein synthesis, whereas the NR2B-containing antagonist Ro25-6981 did not. Conclusions PCP dysregulates translation, acting through NR2A-containing NMDAR subtypes, recruiting mGluR1/5 signaling pathways, and leading to neural discoordination that is central to the cognitive and sensorimotor impairments.
Collapse
Affiliation(s)
- Eun Hye Park
- Center for Neural Science, New York University, New York, New York
| | - Hsin-Yi Kao
- Center for Neural Science, New York University, New York, New York
| | - Hussam Jourdi
- Center for Neural Science, New York University, New York, New York
| | - Milenna T. van Dijk
- Center for Neural Science, New York University, New York, New York
- Graduate Program in Neuroscience and Physiology, New York University Langone Medical Center, New York, New York
| | - Simón Carrillo-Segura
- Center for Neural Science, New York University, New York, New York
- Graduate Program in Mechanical and Aerospace Engineering, New York University Tandon School of Engineering, New York, New York
| | - Kayla W. Tunnell
- Center for Neural Science, New York University, New York, New York
| | | | - Emma J. Wallace
- Graduate Program in Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Department of Physiology and Pharmacology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - Matthew Troy-Regier
- Graduate Program in Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Department of Physiology and Pharmacology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - Basma Radwan
- Graduate Program in Neural Science, Center for Neural Science, New York University, New York, New York
| | | | - Juan Marcos Alarcon
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - André A. Fenton
- Center for Neural Science, New York University, New York, New York
- Department of Physiology and Pharmacology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, New York
- Neuroscience Institute, NYU Langone Health, New York, New York
| |
Collapse
|
8
|
Provenzano F, Torazza C, Bonifacino T, Bonanno G, Milanese M. The Key Role of Astrocytes in Amyotrophic Lateral Sclerosis and Their Commitment to Glutamate Excitotoxicity. Int J Mol Sci 2023; 24:15430. [PMID: 37895110 PMCID: PMC10607805 DOI: 10.3390/ijms242015430] [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: 08/25/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
In the last two decades, there has been increasing evidence supporting non-neuronal cells as active contributors to neurodegenerative disorders. Among glial cells, astrocytes play a pivotal role in driving amyotrophic lateral sclerosis (ALS) progression, leading the scientific community to focus on the "astrocytic signature" in ALS. Here, we summarized the main pathological mechanisms characterizing astrocyte contribution to MN damage and ALS progression, such as neuroinflammation, mitochondrial dysfunction, oxidative stress, energy metabolism impairment, miRNAs and extracellular vesicles contribution, autophagy dysfunction, protein misfolding, and altered neurotrophic factor release. Since glutamate excitotoxicity is one of the most relevant ALS features, we focused on the specific contribution of ALS astrocytes in this aspect, highlighting the known or potential molecular mechanisms by which astrocytes participate in increasing the extracellular glutamate level in ALS and, conversely, undergo the toxic effect of the excessive glutamate. In this scenario, astrocytes can behave as "producers" and "targets" of the high extracellular glutamate levels, going through changes that can affect themselves and, in turn, the neuronal and non-neuronal surrounding cells, thus actively impacting the ALS course. Moreover, this review aims to point out knowledge gaps that deserve further investigation.
Collapse
Affiliation(s)
- Francesca Provenzano
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
| | - Carola Torazza
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
| | - Tiziana Bonifacino
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
| | - Marco Milanese
- Department of Pharmacy (DIFAR), University of Genoa, 16148 Genova, Italy; (F.P.); (C.T.); (G.B.); (M.M.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
9
|
de Ceglia R, Ledonne A, Litvin DG, Lind BL, Carriero G, Latagliata EC, Bindocci E, Di Castro MA, Savtchouk I, Vitali I, Ranjak A, Congiu M, Canonica T, Wisden W, Harris K, Mameli M, Mercuri N, Telley L, Volterra A. Specialized astrocytes mediate glutamatergic gliotransmission in the CNS. Nature 2023; 622:120-129. [PMID: 37674083 PMCID: PMC10550825 DOI: 10.1038/s41586-023-06502-w] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 07/31/2023] [Indexed: 09/08/2023]
Abstract
Multimodal astrocyte-neuron communications govern brain circuitry assembly and function1. For example, through rapid glutamate release, astrocytes can control excitability, plasticity and synchronous activity2,3 of synaptic networks, while also contributing to their dysregulation in neuropsychiatric conditions4-7. For astrocytes to communicate through fast focal glutamate release, they should possess an apparatus for Ca2+-dependent exocytosis similar to neurons8-10. However, the existence of this mechanism has been questioned11-13 owing to inconsistent data14-17 and a lack of direct supporting evidence. Here we revisited the astrocyte glutamate exocytosis hypothesis by considering the emerging molecular heterogeneity of astrocytes18-21 and using molecular, bioinformatic and imaging approaches, together with cell-specific genetic tools that interfere with glutamate exocytosis in vivo. By analysing existing single-cell RNA-sequencing databases and our patch-seq data, we identified nine molecularly distinct clusters of hippocampal astrocytes, among which we found a notable subpopulation that selectively expressed synaptic-like glutamate-release machinery and localized to discrete hippocampal sites. Using GluSnFR-based glutamate imaging22 in situ and in vivo, we identified a corresponding astrocyte subgroup that responds reliably to astrocyte-selective stimulations with subsecond glutamate release events at spatially precise hotspots, which were suppressed by astrocyte-targeted deletion of vesicular glutamate transporter 1 (VGLUT1). Furthermore, deletion of this transporter or its isoform VGLUT2 revealed specific contributions of glutamatergic astrocytes in cortico-hippocampal and nigrostriatal circuits during normal behaviour and pathological processes. By uncovering this atypical subpopulation of specialized astrocytes in the adult brain, we provide insights into the complex roles of astrocytes in central nervous system (CNS) physiology and diseases, and identify a potential therapeutic target.
Collapse
Affiliation(s)
- Roberta de Ceglia
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Ada Ledonne
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
- Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy
| | - David Gregory Litvin
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
- Wyss Center for Bio and Neuro Engineering, Campus Biotech, Geneva, Switzerland
| | - Barbara Lykke Lind
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Giovanni Carriero
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | | | - Erika Bindocci
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | | | - Iaroslav Savtchouk
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, USA
| | - Ilaria Vitali
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Anurag Ranjak
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Mauro Congiu
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Tara Canonica
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - William Wisden
- Department of Life Sciences and UK Dementia Research Institute, Imperial College London, London, UK
| | - Kenneth Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Manuel Mameli
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Nicola Mercuri
- Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Ludovic Telley
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland.
| | - Andrea Volterra
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland.
- Wyss Center for Bio and Neuro Engineering, Campus Biotech, Geneva, Switzerland.
| |
Collapse
|
10
|
Pereira MJ, Ayana R, Holt MG, Arckens L. Chemogenetic manipulation of astrocyte activity at the synapse- a gateway to manage brain disease. Front Cell Dev Biol 2023; 11:1193130. [PMID: 37534103 PMCID: PMC10393042 DOI: 10.3389/fcell.2023.1193130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 08/04/2023] Open
Abstract
Astrocytes are the major glial cell type in the central nervous system (CNS). Initially regarded as supportive cells, it is now recognized that this highly heterogeneous cell population is an indispensable modulator of brain development and function. Astrocytes secrete neuroactive molecules that regulate synapse formation and maturation. They also express hundreds of G protein-coupled receptors (GPCRs) that, once activated by neurotransmitters, trigger intracellular signalling pathways that can trigger the release of gliotransmitters which, in turn, modulate synaptic transmission and neuroplasticity. Considering this, it is not surprising that astrocytic dysfunction, leading to synaptic impairment, is consistently described as a factor in brain diseases, whether they emerge early or late in life due to genetic or environmental factors. Here, we provide an overview of the literature showing that activation of genetically engineered GPCRs, known as Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), to specifically modulate astrocyte activity partially mimics endogenous signalling pathways in astrocytes and improves neuronal function and behavior in normal animals and disease models. Therefore, we propose that expressing these genetically engineered GPCRs in astrocytes could be a promising strategy to explore (new) signalling pathways which can be used to manage brain disorders. The precise molecular, functional and behavioral effects of this type of manipulation, however, differ depending on the DREADD receptor used, targeted brain region and timing of the intervention, between healthy and disease conditions. This is likely a reflection of regional and disease/disease progression-associated astrocyte heterogeneity. Therefore, a thorough investigation of the effects of such astrocyte manipulation(s) must be conducted considering the specific cellular and molecular environment characteristic of each disease and disease stage before this has therapeutic applicability.
Collapse
Affiliation(s)
- Maria João Pereira
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Rajagopal Ayana
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Matthew G. Holt
- Instituto de Investigação e Inovação em Saúde (i3S), Laboratory of Synapse Biology, Universidade do Porto, Porto, Portugal
| | - Lutgarde Arckens
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
11
|
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: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [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
|
12
|
Lee SH, Mak A, Verheijen MHG. Comparative assessment of the effects of DREADDs and endogenously expressed GPCRs in hippocampal astrocytes on synaptic activity and memory. Front Cell Neurosci 2023; 17:1159756. [PMID: 37051110 PMCID: PMC10083367 DOI: 10.3389/fncel.2023.1159756] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) have proven themselves as one of the key in vivo techniques of modern neuroscience, allowing for unprecedented access to cellular manipulations in living animals. With respect to astrocyte research, DREADDs have become a popular method to examine the functional aspects of astrocyte activity, particularly G-protein coupled receptor (GPCR)-mediated intracellular calcium (Ca2+) and cyclic adenosine monophosphate (cAMP) dynamics. With this method it has become possible to directly link the physiological aspects of astrocytic function to cognitive processes such as memory. As a result, a multitude of studies have explored the impact of DREADD activation in astrocytes on synaptic activity and memory. However, the emergence of varying results prompts us to reconsider the degree to which DREADDs expressed in astrocytes accurately mimic endogenous GPCR activity. Here we compare the major downstream signaling mechanisms, synaptic, and behavioral effects of stimulating Gq-, Gs-, and Gi-DREADDs in hippocampal astrocytes of adult mice to those of endogenously expressed GPCRs.
Collapse
Affiliation(s)
- Sophie H. Lee
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Research Master’s Programme Brain and Cognitive Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Aline Mak
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mark H. G. Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: Mark Verheijen,
| |
Collapse
|
13
|
Mughal A, Nelson MT, Hill-Eubanks D. The post-arteriole transitional zone: a specialized capillary region that regulates blood flow within the CNS microvasculature. J Physiol 2023; 601:889-901. [PMID: 36751860 PMCID: PMC9992301 DOI: 10.1113/jp282246] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023] Open
Abstract
The brain is an energy hog, consuming available energy supplies at a rate out of all proportion to its relatively small size. This outsized demand, largely reflecting the unique computational activity of the brain, is met by an ensemble of neurovascular coupling mechanisms that link neuronal activity with local increases in blood delivery. This just-in-time replenishment strategy, made necessary by the limited energy-storage capacity of neurons, complicates the nutrient-delivery task of the cerebral vasculature, layering on a temporo-spatial requirement that invites - and challenges - mechanistic interpretation. The centre of gravity of research efforts to disentangle these mechanisms has shifted from an initial emphasis on astrocyte-arteriole-level processes to mechanisms that operate on the capillary level, a shift that has brought into sharp focus questions regarding the fine control of blood distribution to active neurons. As these investigations have drilled down into finer reaches of the microvasculature, they have revealed an arteriole-proximate subregion of CNS capillary networks that serves a regulatory function in directing blood flow into and within downstream capillaries. They have also illuminated differences in researchers' perspectives on the vascular structures and identity of mural cells in this region that impart the vasomodulatory effects that control blood distribution. In this review, we highlight the regulatory role of a variably named region of the microvasculature, referred to here as the post-arteriole transition zone, in channeling blood flow within CNS capillary networks, and underscore the contribution of dynamically contractile perivascular mural cell - generally, but not universally, recognized as pericytes - to this function.
Collapse
Affiliation(s)
- Amreen Mughal
- Department of Pharmacology, University of Vermont, Burlington, VT, USA
| | - Mark T. Nelson
- Department of Pharmacology, University of Vermont, Burlington, VT, USA
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | | |
Collapse
|
14
|
Astrocytic Piezo1-mediated mechanotransduction determines adult neurogenesis and cognitive functions. Neuron 2022; 110:2984-2999.e8. [PMID: 35963237 DOI: 10.1016/j.neuron.2022.07.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 05/31/2022] [Accepted: 07/12/2022] [Indexed: 12/12/2022]
Abstract
Adult brain activities are generally believed to be dominated by chemical and electrical transduction mechanisms. However, the importance of mechanotransduction mediated by mechano-gated ion channels in brain functions is less appreciated. Here, we show that the mechano-gated Piezo1 channel is expressed in the exploratory processes of astrocytes and utilizes its mechanosensitivity to mediate mechanically evoked Ca2+ responses and ATP release, establishing Piezo1-mediated mechano-chemo transduction in astrocytes. Piezo1 deletion in astrocytes causes a striking reduction of hippocampal volume and brain weight and severely impaired (but ATP-rescuable) adult neurogenesis in vivo, and it abolishes ATP-dependent potentiation of neural stem cell (NSC) proliferation in vitro. Piezo1-deficient mice show impaired hippocampal long-term potentiation (LTP) and learning and memory behaviors. By contrast, overexpression of Piezo1 in astrocytes sufficiently enhances mechanotransduction, LTP, and learning and memory performance. Thus, astrocytes utilize Piezo1-mediated mechanotransduction mechanisms to robustly regulate adult neurogenesis and cognitive functions, conceptually highlighting the importance of mechanotransduction in brain structure and function.
Collapse
|
15
|
Walch E, Bilas A, Bebawy V, Lam A, Murphy TR, Sriram S, Fiacco TA. Contributions of Astrocyte and Neuronal Volume to CA1 Neuron Excitability Changes in Elevated Extracellular Potassium. Front Cell Neurosci 2022; 16:930384. [PMID: 35936495 PMCID: PMC9352931 DOI: 10.3389/fncel.2022.930384] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Rapid increases in cell volume reduce the size of the extracellular space (ECS) and are associated with elevated brain tissue excitability. We recently demonstrated that astrocytes, but not neurons, rapidly swell in elevated extracellular potassium (∧[K+]o) up to 26 mM. However, effects of acute astrocyte volume fluctuations on neuronal excitability in ∧[K+]o have been difficult to evaluate due to direct effects on neuronal membrane potential and generation of action potentials. Here we set out to isolate volume-specific effects occurring in ∧[K+]o on CA1 pyramidal neurons in acute hippocampal slices by manipulating cell volume while recording neuronal glutamate currents in 10.5 mM [K+]o + tetrodotoxin (TTX) to prevent neuronal firing. Elevating [K+]o to 10.5 mM induced astrocyte swelling and produced significant increases in neuronal excitability in the form of mixed α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/N-methyl-D-aspartate (NMDA) receptor mEPSCs and NMDA receptor-dependent slow inward currents (SICs). Application of hyperosmolar artificial cerebrospinal fluid (ACSF) by addition of mannitol in the continued presence of 10.5 mM K+ forced shrinking of astrocytes and to a lesser extent neurons, which resisted swelling in ∧[K+]o. Cell shrinking and dilation of the ECS significantly dampened neuronal excitability in 10.5 mM K+. Subsequent removal of mannitol amplified effects on neuronal excitability and nearly doubled the volume increase in astrocytes, presumably due to continued glial uptake of K+ while mannitol was present. Slower, larger amplitude events mainly driven by NMDA receptors were abolished by mannitol-induced expansion of the ECS. Collectively, our findings suggest that cell volume regulation of the ECS in elevated [K+]o is driven predominantly by astrocytes, and that cell volume effects on neuronal excitability can be effectively isolated in elevated [K+]o conditions.
Collapse
Affiliation(s)
- Erin Walch
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
- Center for Glial-Neuronal Interactions, University of California, Riverside, Riverside, CA, United States
| | - Alexander Bilas
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
| | - Valine Bebawy
- Undergraduate Major in Biology, University of California, Riverside, Riverside, CA, United States
| | - Angelina Lam
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Thomas R. Murphy
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
- Center for Glial-Neuronal Interactions, University of California, Riverside, Riverside, CA, United States
| | - Sandhya Sriram
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
| | - Todd A. Fiacco
- Center for Glial-Neuronal Interactions, University of California, Riverside, Riverside, CA, United States
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
- *Correspondence: Todd A. Fiacco,
| |
Collapse
|
16
|
Satarker S, Bojja SL, Gurram PC, Mudgal J, Arora D, Nampoothiri M. Astrocytic Glutamatergic Transmission and Its Implications in Neurodegenerative Disorders. Cells 2022; 11:cells11071139. [PMID: 35406702 PMCID: PMC8997779 DOI: 10.3390/cells11071139] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/12/2022] [Accepted: 03/13/2022] [Indexed: 12/11/2022] Open
Abstract
Several neurodegenerative disorders involve impaired neurotransmission, and glutamatergic neurotransmission sets a prototypical example. Glutamate is a predominant excitatory neurotransmitter where the astrocytes play a pivotal role in maintaining the extracellular levels through release and uptake mechanisms. Astrocytes modulate calcium-mediated excitability and release several neurotransmitters and neuromodulators, including glutamate, and significantly modulate neurotransmission. Accumulating evidence supports the concept of excitotoxicity caused by astrocytic glutamatergic release in pathological conditions. Thus, the current review highlights different vesicular and non-vesicular mechanisms of astrocytic glutamate release and their implication in neurodegenerative diseases. As in presynaptic neurons, the vesicular release of astrocytic glutamate is also primarily meditated by calcium-mediated exocytosis. V-ATPase is crucial in the acidification and maintenance of the gradient that facilitates the vesicular storage of glutamate. Along with these, several other components, such as cystine/glutamate antiporter, hemichannels, BEST-1, TREK-1, purinergic receptors and so forth, also contribute to glutamate release under physiological and pathological conditions. Events of hampered glutamate uptake could promote inflamed astrocytes to trigger repetitive release of glutamate. This could be favorable towards the development and worsening of neurodegenerative diseases. Therefore, across neurodegenerative diseases, we review the relations between defective glutamatergic signaling and astrocytic vesicular and non-vesicular events in glutamate homeostasis. The optimum regulation of astrocytic glutamatergic transmission could pave the way for the management of these diseases and add to their therapeutic value.
Collapse
Affiliation(s)
- Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.S.); (S.L.B.); (P.C.G.); (J.M.)
| | - Sree Lalitha Bojja
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.S.); (S.L.B.); (P.C.G.); (J.M.)
| | - Prasada Chowdari Gurram
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.S.); (S.L.B.); (P.C.G.); (J.M.)
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.S.); (S.L.B.); (P.C.G.); (J.M.)
| | - Devinder Arora
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.S.); (S.L.B.); (P.C.G.); (J.M.)
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.S.); (S.L.B.); (P.C.G.); (J.M.)
- Correspondence:
| |
Collapse
|
17
|
Distinct roles of astroglia and neurons in synaptic plasticity and memory. Mol Psychiatry 2022; 27:873-885. [PMID: 34642458 DOI: 10.1038/s41380-021-01332-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/17/2021] [Accepted: 09/29/2021] [Indexed: 12/17/2022]
Abstract
Long-term potentiation (LTP) in the hippocampus is the most studied form of synaptic plasticity. Temporal integration of synaptic inputs is essential in synaptic plasticity and is assumed to be achieved through Ca2+ signaling in neurons and astroglia. However, whether these two cell types play different roles in LTP remain unknown. Here, we found that through the integration of synaptic inputs, astrocyte inositol triphosphate (IP3) receptor type 2 (IP3R2)-dependent Ca2+ signaling was critical for late-phase LTP (L-LTP) but not early-phase LTP (E-LTP). Moreover, this process was mediated by astrocyte-derived brain-derived neurotrophic factor (BDNF). In contrast, neuron-derived BDNF was critical for both E-LTP and L-LTP. Importantly, the dynamic differences in BDNF secretion play a role in modulating distinct forms of LTP. Moreover, astrocyte- and neuron-derived BDNF exhibited different roles in memory. These observations enriched our knowledge of LTP and memory at the cellular level and implied distinct roles of astrocytes and neurons in information integration.
Collapse
|
18
|
Abstract
Drug addiction remains a key biomedical challenge facing current neuroscience research. In addition to neural mechanisms, the focus of the vast majority of studies to date, astrocytes have been increasingly recognized as an "accomplice." According to the tripartite synapse model, astrocytes critically regulate nearby pre- and postsynaptic neuronal substrates to craft experience-dependent synaptic plasticity, including synapse formation and elimination. Astrocytes within brain regions that are implicated in drug addiction exhibit dynamic changes in activity upon exposure to cocaine and subsequently undergo adaptive changes themselves during chronic drug exposure. Recent results have identified several key astrocytic signaling pathways that are involved in cocaine-induced synaptic and circuit adaptations. In this review, we provide a brief overview of the role of astrocytes in regulating synaptic transmission and neuronal function, and discuss how cocaine influences these astrocyte-mediated mechanisms to induce persistent synaptic and circuit alterations that promote cocaine seeking and relapse. We also consider the therapeutic potential of targeting astrocytic substrates to ameliorate drug-induced neuroplasticity for behavioral benefits. While primarily focusing on cocaine-induced astrocytic responses, we also include brief discussion of other drugs of abuse where data are available.
Collapse
|
19
|
O'Donovan B, Neugornet A, Neogi R, Xia M, Ortinski P. Cocaine experience induces functional adaptations in astrocytes: Implications for synaptic plasticity in the nucleus accumbens shell. Addict Biol 2021; 26:e13042. [PMID: 33864336 DOI: 10.1111/adb.13042] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 11/24/2022]
Abstract
Astrocytes have become established as an important regulator of neuronal activity in the brain. Accumulating literature demonstrates that cocaine self-administration in rodent models induces structural changes within astrocytes that may influence their interaction with the surrounding neurons. Here, we provide evidence that cocaine impacts astrocytes at the functional level and alters neuronal sensitivity to astrocyte-derived glutamate. We report that a 14-day period of short access to cocaine (2 h/day) decreases spontaneous astrocytic Ca2+ transients and precipitates changes in astrocyte network activity in the nucleus accumbens shell. This is accompanied by increased prevalence of slow inward currents, a physiological marker of neuronal activation by astrocytic glutamate, in a subset of medium spiny neurons. Within, but not outside, of this subset, we observe an increase in duration and frequency of N-methyl-D-aspartate (NMDA) receptor-mediated synaptic events. Additionally, we find that the link between synaptic NMDA receptor plasticity and neuron sensitivity to astrocytic glutamate is maintained independent of drug exposure and is observed in both cocaine and saline control animals. Imaging analyses of neuronal Ca2+ activity show no effect of cocaine self-administration on individual cells or on neuronal network activity in brain slices. Therefore, our data indicate that cocaine self-administration promotes astrocyte-specific functional changes that can be linked to increased glutamate-mediated coupling with principal neurons in the nucleus accumbens. Such coupling may be spatially restricted as it does not result in a broad impact on network structure of local neuronal circuits.
Collapse
Affiliation(s)
- Bernadette O'Donovan
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| | - Austin Neugornet
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| | - Richik Neogi
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
- Integrated Biomedical Sciences University of Kentucky Lexington Kentucky USA
| | - Mengfan Xia
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| | - Pavel Ortinski
- Department of Neuroscience, College of Medicine University of Kentucky Lexington Kentucky USA
| |
Collapse
|
20
|
Sherwood MW, Arizono M, Panatier A, Mikoshiba K, Oliet SHR. Astrocytic IP 3Rs: Beyond IP 3R2. Front Cell Neurosci 2021; 15:695817. [PMID: 34393726 PMCID: PMC8363081 DOI: 10.3389/fncel.2021.695817] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/30/2021] [Indexed: 12/31/2022] Open
Abstract
Astrocytes are sensitive to ongoing neuronal/network activities and, accordingly, regulate neuronal functions (synaptic transmission, synaptic plasticity, behavior, etc.) by the context-dependent release of several gliotransmitters (e.g., glutamate, glycine, D-serine, ATP). To sense diverse input, astrocytes express a plethora of G-protein coupled receptors, which couple, via Gi/o and Gq, to the intracellular Ca2+ release channel IP3-receptor (IP3R). Indeed, manipulating astrocytic IP3R-Ca2+ signaling is highly consequential at the network and behavioral level: Depleting IP3R subtype 2 (IP3R2) results in reduced GPCR-Ca2+ signaling and impaired synaptic plasticity; enhancing IP3R-Ca2+ signaling affects cognitive functions such as learning and memory, sleep, and mood. However, as a result of discrepancies in the literature, the role of GPCR-IP3R-Ca2+ signaling, especially under physiological conditions, remains inconclusive. One primary reason for this could be that IP3R2 has been used to represent all astrocytic IP3Rs, including IP3R1 and IP3R3. Indeed, IP3R1 and IP3R3 are unique Ca2+ channels in their own right; they have unique biophysical properties, often display distinct distribution, and are differentially regulated. As a result, they mediate different physiological roles to IP3R2. Thus, these additional channels promise to enrich the diversity of spatiotemporal Ca2+ dynamics and provide unique opportunities for integrating neuronal input and modulating astrocyte–neuron communication. The current review weighs evidence supporting the existence of multiple astrocytic-IP3R isoforms, summarizes distinct sub-type specific properties that shape spatiotemporal Ca2+ dynamics. We also discuss existing experimental tools and future refinements to better recapitulate the endogenous activities of each IP3R isoform.
Collapse
Affiliation(s)
- Mark W Sherwood
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Misa Arizono
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Aude Panatier
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Katsuhiko Mikoshiba
- ShanghaiTech University, Shanghai, China.,Faculty of Science, Toho University, Funabashi, Japan.,RIKEN CLST, Kobe, Japan
| | - Stéphane H R Oliet
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| |
Collapse
|
21
|
Lim EY, Ye L, Paukert M. Potential and Realized Impact of Astroglia Ca 2 + Dynamics on Circuit Function and Behavior. Front Cell Neurosci 2021; 15:682888. [PMID: 34163330 PMCID: PMC8215280 DOI: 10.3389/fncel.2021.682888] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/03/2021] [Indexed: 11/13/2022] Open
Abstract
Astroglia display a wide range of spontaneous and behavioral state-dependent Ca2+ dynamics. During heightened vigilance, noradrenergic signaling leads to quasi-synchronous Ca2+ elevations encompassing soma and processes across the brain-wide astroglia network. Distinct from this vigilance-associated global Ca2+ rise are apparently spontaneous fluctuations within spatially restricted microdomains. Over the years, several strategies have been pursued to shed light on the physiological impact of these signals including deletion of endogenous ion channels or receptors and reduction of intracellular Ca2+ through buffering, extrusion or inhibition of release. Some experiments that revealed the most compelling behavioral alterations employed chemogenetic and optogenetic manipulations to modify astroglia Ca2+ signaling. However, there is considerable contrast between these findings and the comparatively modest effects of inhibiting endogenous sources of Ca2+. In this review, we describe the underlying mechanisms of various forms of astroglia Ca2+ signaling as well as the functional consequences of their inhibition. We then discuss how the effects of exogenous astroglia Ca2+ modification combined with our knowledge of physiological mechanisms of astroglia Ca2+ activation could guide further refinement of behavioral paradigms that will help elucidate the natural Ca2+-dependent function of astroglia.
Collapse
Affiliation(s)
- Eunice Y. Lim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Liang Ye
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Martin Paukert
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States,*Correspondence: Martin Paukert,
| |
Collapse
|
22
|
Clyburn C, Browning KN. Glutamatergic plasticity within neurocircuits of the dorsal vagal complex and the regulation of gastric functions. Am J Physiol Gastrointest Liver Physiol 2021; 320:G880-G887. [PMID: 33730858 PMCID: PMC8202199 DOI: 10.1152/ajpgi.00014.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The meticulous regulation of the gastrointestinal (GI) tract is required for the coordination of gastric motility and emptying, intestinal secretion, absorption, and transit as well as for the overarching management of food intake and energy homeostasis. Disruption of GI functions is associated with the development of severe GI disorders and the alteration of food intake and caloric balance. Functional GI disorders as well as the dysregulation of energy balance and food intake are frequently associated with, or result from, alterations in the central regulation of GI control. The faithful and rapid transmission of information from the stomach and upper GI tract to second-order neurons of the nucleus of the tractus solitarius (NTS) relies on the delicate modulation of excitatory glutamatergic transmission, as does the relay of integrated signals from the NTS to parasympathetic efferent neurons of the dorsal motor nucleus of the vagus (DMV). Many studies have focused on understanding the physiological and pathophysiological modulation of these glutamatergic synapses, although their role in the control and regulation of GI functions has lagged behind that of cardiovascular and respiratory functions. The purpose of this review is to examine the current literature exploring the role of glutamatergic transmission in the DVC in the regulation of GI functions.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
23
|
Broadhead MJ, Miles GB. A common role for astrocytes in rhythmic behaviours? Prog Neurobiol 2021; 202:102052. [PMID: 33894330 DOI: 10.1016/j.pneurobio.2021.102052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/03/2021] [Accepted: 04/13/2021] [Indexed: 01/16/2023]
Abstract
Astrocytes are a functionally diverse form of glial cell involved in various aspects of nervous system infrastructure, from the metabolic and structural support of neurons to direct neuromodulation of synaptic activity. Investigating how astrocytes behave in functionally related circuits may help us understand whether there is any conserved logic to the role of astrocytes within neuronal networks. Astrocytes are implicated as key neuromodulatory cells within neural circuits that control a number of rhythmic behaviours such as breathing, locomotion and circadian sleep-wake cycles. In this review, we examine the evidence that astrocytes are directly involved in the regulation of the neural circuits underlying six different rhythmic behaviours: locomotion, breathing, chewing, gastrointestinal motility, circadian sleep-wake cycles and oscillatory feeding behaviour. We discuss how astrocytes are integrated into the neuronal networks that regulate these behaviours, and identify the potential gliotransmission signalling mechanisms involved. From reviewing the evidence of astrocytic involvement in a range of rhythmic behaviours, we reveal a heterogenous array of gliotransmission mechanisms, which help to regulate neuronal networks. However, we also observe an intriguing thread of commonality, in the form of purinergic gliotransmission, which is frequently utilised to facilitate feedback inhibition within rhythmic networks to constrain a given behaviour within its operational range.
Collapse
Affiliation(s)
- Matthew J Broadhead
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK.
| | - Gareth B Miles
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| |
Collapse
|
24
|
Chemogenetic manipulation of astrocytic activity: Is it possible to reveal the roles of astrocytes? Biochem Pharmacol 2021; 186:114457. [PMID: 33556341 DOI: 10.1016/j.bcp.2021.114457] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 01/08/2023]
Abstract
Astrocytes are the major glial cells in the central nervous system, but unlike neurons, they do not produce action potentials. For many years, astrocytes were considered supporting cells in the central nervous system (CNS). Technological advances over the last two decades are changing the face of glial research. Accumulating data from recent investigations show that astrocytes display transient calcium spikes and regulate synaptic transmission by releasing transmitters called gliotransmitters. Many new powerful technologies are used to interfere with astrocytic activity, in order to obtain a better understanding of the roles of astrocytes in the brain. Among these technologies, chemogenetics has recently been used frequently. In this review, we will summarize new functions of astrocytes in the brain that have been revealed using this cutting-edge technique. Moreover, we will discuss the possibilities and challenges of manipulating astrocytic activity using this technology.
Collapse
|
25
|
Guillot de Suduiraut I, Grosse J, Ramos-Fernández E, Sandi C, Hollis F. Astrocytic release of ATP through type 2 inositol 1,4,5-trisphosphate receptor calcium signaling and social dominance behavior in mice. Eur J Neurosci 2020; 53:2973-2985. [PMID: 32609904 DOI: 10.1111/ejn.14892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/22/2020] [Accepted: 06/18/2020] [Indexed: 01/08/2023]
Abstract
Brain mitochondrial function is critical for numerous neuronal processes. We recently identified a link between brain energy and social dominance, where higher levels of mitochondrial function resulted in increased social competitive ability. The underlying mechanism of this link, however, remains unclear. Here, we investigated the contribution of astrocytic release of adenosine triphosphate (ATP) through the type 2 inositol 1,4,5-triphosphate receptor to social dominance behavior. Mice lacking the type 2 inositol 1,4,5-triphosphate receptor were characterized for their social dominance behavior, as well as their performance on a nonsocial task, the Morris Water Maze. In parallel, we also examined mitochondrial function in the medial prefrontal cortex, nucleus accumbens, and hippocampus to investigate how deficiencies in astrocytic ATP could modulate overall mitochondrial function. While knockout mice showed similar competitive ability compared with their wild-type littermates, dominant knockout mice exhibited a significant delay in exerting their dominance during the initial encounter. Otherwise, there were no differences in anxiety and exploratory traits, spatial learning and memory, or brain mitochondrial function in either light or dark circadian phases. Our findings point to a marginal role of astrocytic ATP through IP3 R2 in social competition, suggesting that, under basal conditions, the neuronal compartment is predominant for social dominance exertion.
Collapse
Affiliation(s)
| | - Jocelyn Grosse
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Eva Ramos-Fernández
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carmen Sandi
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Fiona Hollis
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia
| |
Collapse
|
26
|
LRRC8A-dependent volume-regulated anion channels contribute to ischemia-induced brain injury and glutamatergic input to hippocampal neurons. Exp Neurol 2020; 332:113391. [PMID: 32598930 DOI: 10.1016/j.expneurol.2020.113391] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/05/2020] [Accepted: 06/25/2020] [Indexed: 11/20/2022]
Abstract
Volume-regulated anion channels (VRACs) are critically involved in regulating cell volume, and leucine-rich repeat-containing protein 8A (LRRC8A, SWELL1) is an obligatory subunit of VRACs. Cell swelling occurs early after brain ischemia, but it is unclear whether neuronal LRRC8a contributes to ischemia-induced glutamate release and brain injury. We found that Lrrc8a conditional knockout (Lrrc8a-cKO) mice produced by crossing NestinCre+/- with Lrrc8aflox+/+ mice died 7-8 weeks of age, indicating an essential role of neuronal LRRC8A for survival. Middle cerebral artery occlusion (MCAO) caused an early increase in LRRC8A protein levels in the hippocampus in wild-type (WT) mice. Whole-cell patch-clamp recording in brain slices revealed that oxygen-glucose deprivation significantly increased the amplitude of VRAC currents in hippocampal CA1 neurons in WT but not in Lrrc8a-cKO mice. Hypotonicity increased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) in hippocampal CA1 neurons in WT mice, and this was abolished by DCPIB, a VRAC blocker. But in Lrrc8a-cKO mice, hypotonic solution had no effect on the frequency of sEPSCs in these neurons. Furthermore, the brain infarct volume and neurological severity score induced by MCAO were significantly lower in Lrrc8a-cKO mice than in WT mice. In addition, MCAO-induced increases in cleaved caspase-3 and calpain activity, two biochemical markers of neuronal apoptosis and death, in brain tissues were significantly attenuated in Lrrc8a-cKO mice compared with WT mice. These new findings indicate that cerebral ischemia increases neuronal LRRC8A-dependent VRAC activity and that VRACs contribute to increased glutamatergic input to hippocampal neurons and brain injury caused by ischemic stroke.
Collapse
|
27
|
Pham C, Moro DH, Mouffle C, Didienne S, Hepp R, Pfrieger FW, Mangin JM, Legendre P, Martin C, Luquet S, Cauli B, Li D. Mapping astrocyte activity domains by light sheet imaging and spatio-temporal correlation screening. Neuroimage 2020; 220:117069. [PMID: 32585347 DOI: 10.1016/j.neuroimage.2020.117069] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 02/08/2023] Open
Abstract
Astrocytes are a major type of glial cell in the mammalian brain, essentially regulating neuronal development and function. Quantitative imaging represents an important approach to study astrocytic signaling in neural circuits. Focusing on astrocytic Ca2+ activity, a key pathway implicated in astrocye-neuron interaction, we here report a strategy combining fast light sheet fluorescence microscopy (LSFM) and correlative screening-based time series analysis, to map activity domains in astrocytes in living mammalian nerve tissue. Light sheet of micron-scale thickness enables wide-field optical sectioning to image astrocytes in acute mouse brain slices. Using both chemical and genetically encoded Ca2+ indicators, we demonstrate the complementary advantages of LSFM in mapping Ca2+ domains in astrocyte populations as compared to epifluorescence and two-photon microscopy. Our approach then revealed distinct kinetics of Ca2+ signals between cortical and hypothalamic astrocytes in resting conditions and following the activation of adrenergic G protein coupled receptor (GPCR). This observation highlights the activity heterogeneity across regionally distinct astrocyte populations, and indicates the potential of our method for investigating dynamic signals in astrocytes.
Collapse
Affiliation(s)
- Cuong Pham
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Daniela Herrera Moro
- Unité de Biologie Fonctionnelle et Adaptative, Centre National la Recherche Scientifique, Unité Mixte de Recherche 8251, Université Paris Diderot, Sorbonne Paris Cité, 75205, Paris, France
| | - Christine Mouffle
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Steve Didienne
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Régine Hepp
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Frank W Pfrieger
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, F-67000, Strasbourg, France
| | - Jean-Marie Mangin
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Pascal Legendre
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Claire Martin
- Unité de Biologie Fonctionnelle et Adaptative, Centre National la Recherche Scientifique, Unité Mixte de Recherche 8251, Université Paris Diderot, Sorbonne Paris Cité, 75205, Paris, France
| | - Serge Luquet
- Unité de Biologie Fonctionnelle et Adaptative, Centre National la Recherche Scientifique, Unité Mixte de Recherche 8251, Université Paris Diderot, Sorbonne Paris Cité, 75205, Paris, France
| | - Bruno Cauli
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France
| | - Dongdong Li
- Sorbonne Université, Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR8246, INSERM U1130, UPMC UMCR18, Paris, 75005, France.
| |
Collapse
|
28
|
Martinez-Banaclocha M. Astroglial Isopotentiality and Calcium-Associated Biomagnetic Field Effects on Cortical Neuronal Coupling. Cells 2020; 9:cells9020439. [PMID: 32069981 PMCID: PMC7073214 DOI: 10.3390/cells9020439] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 01/01/2023] Open
Abstract
Synaptic neurotransmission is necessary but does not sufficiently explain superior cognitive faculties. Growing evidence has shown that neuron-astroglial chemical crosstalk plays a critical role in the processing of information, computation, and memory. In addition to chemical and electrical communication among neurons and between neurons and astrocytes, other nonsynaptic mechanisms called ephaptic interactions can contribute to the neuronal synchronization from different brain regions involved in the processing of information. New research on brain astrocytes has clearly shown that the membrane potential of these cells remains very stable among neighboring and distant astrocytes due to the marked bioelectric coupling between them through gap junctions. This finding raises the possibility that the neocortical astroglial network exerts a guiding template modulating the excitability and synchronization of trillions of neurons by astroglial Ca2+-associated bioelectromagnetic interactions. We propose that bioelectric and biomagnetic fields of the astroglial network equalize extracellular local field potentials (LFPs) and associated local magnetic field potentials (LMFPs) in the cortical layers of the brain areas involved in the processing of information, contributing to the adequate and coherent integration of external and internal signals. This article reviews the current knowledge of ephaptic interactions in the cerebral cortex and proposes that the isopotentiality of cortical astrocytes is a prerequisite for the maintenance of the bioelectromagnetic crosstalk between neurons and astrocytes in the neocortex.
Collapse
|
29
|
Rogers RC, Burke SJ, Collier JJ, Ritter S, Hermann GE. Evidence that hindbrain astrocytes in the rat detect low glucose with a glucose transporter 2-phospholipase C-calcium release mechanism. Am J Physiol Regul Integr Comp Physiol 2020; 318:R38-R48. [PMID: 31596114 PMCID: PMC6985801 DOI: 10.1152/ajpregu.00133.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Astrocytes generate robust cytoplasmic calcium signals in response to reductions in extracellular glucose. This calcium signal, in turn, drives purinergic gliotransmission, which controls the activity of catecholaminergic (CA) neurons in the hindbrain. These CA neurons are critical to triggering glucose counter-regulatory responses (CRRs) that, ultimately, restore glucose homeostasis via endocrine and behavioral means. Although the astrocyte low-glucose sensor involvement in CRR has been accepted, it is not clear how astrocytes produce an increase in intracellular calcium in response to a decrease in glucose. Our ex vivo calcium imaging studies of hindbrain astrocytes show that the glucose type 2 transporter (GLUT2) is an essential feature of the astrocyte glucosensor mechanism. Coimmunoprecipitation assays reveal that the recombinant GLUT2 binds directly with the recombinant Gq protein subunit that activates phospholipase C (PLC). Additional calcium imaging studies suggest that GLUT2 may be connected to a PLC-endoplasmic reticular-calcium release mechanism, which is amplified by calcium-induced calcium release (CICR). Collectively, these data help outline a potential mechanism used by astrocytes to convert information regarding low-glucose levels into intracellular changes that ultimately regulate the CRR.
Collapse
Affiliation(s)
- Richard C. Rogers
- 1Laboratory of Autonomic Neuroscience, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Susan J. Burke
- 2Laboratory of Immunogenetics, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - J. Jason Collier
- 3Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Sue Ritter
- 4Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, Washington State University, Pullman, Washington
| | - Gerlinda E. Hermann
- 1Laboratory of Autonomic Neuroscience, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| |
Collapse
|
30
|
Buskila Y, Bellot-Saez A, Morley JW. Generating Brain Waves, the Power of Astrocytes. Front Neurosci 2019; 13:1125. [PMID: 31680846 PMCID: PMC6813784 DOI: 10.3389/fnins.2019.01125] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022] Open
Abstract
Synchronization of neuronal activity in the brain underlies the emergence of neuronal oscillations termed “brain waves”, which serve various physiological functions and correlate with different behavioral states. It has been postulated that at least ten distinct mechanisms are involved in the formulation of these brain waves, including variations in the concentration of extracellular neurotransmitters and ions, as well as changes in cellular excitability. In this mini review we highlight the contribution of astrocytes, a subtype of glia, in the formation and modulation of brain waves mainly due to their close association with synapses that allows their bidirectional interaction with neurons, and their syncytium-like activity via gap junctions that facilitate communication to distal brain regions through Ca2+ waves. These capabilities allow astrocytes to regulate neuronal excitability via glutamate uptake, gliotransmission and tight control of the extracellular K+ levels via a process termed K+ clearance. Spatio-temporal synchrony of activity across neuronal and astrocytic networks, both locally and distributed across cortical regions, underpins brain states and thereby behavioral states, and it is becoming apparent that astrocytes play an important role in the development and maintenance of neural activity underlying these complex behavioral states.
Collapse
Affiliation(s)
- Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,International Centre for Neuromorphic Systems, The MARCS Institute, Western Sydney University, Penrith, NSW, Australia
| | - Alba Bellot-Saez
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,International Centre for Neuromorphic Systems, The MARCS Institute, Western Sydney University, Penrith, NSW, Australia
| | - John W Morley
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
31
|
Dallérac G, Zapata J, Rouach N. Versatile control of synaptic circuits by astrocytes: where, when and how? Nat Rev Neurosci 2019; 19:729-743. [PMID: 30401802 DOI: 10.1038/s41583-018-0080-6] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Close structural and functional interactions of astrocytes with synapses play an important role in brain function. The repertoire of ways in which astrocytes can regulate synaptic transmission is complex so that they can both promote and dampen synaptic efficacy. Such contrasting effects raise questions regarding the determinants of these divergent astroglial functions. Recent findings provide insights into where, when and how astroglial regulation of synapses takes place by revealing major molecular and functional intrinsic heterogeneity as well as switches in astrocytes occurring during development or specific patterns of neuronal activity. Astrocytes may therefore be seen as boosters or gatekeepers of synaptic circuits depending on their intrinsic and transformative properties throughout life.
Collapse
Affiliation(s)
- Glenn Dallérac
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Jonathan Zapata
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France.
| |
Collapse
|
32
|
Toth AB, Hori K, Novakovic MM, Bernstein NG, Lambot L, Prakriya M. CRAC channels regulate astrocyte Ca 2+ signaling and gliotransmitter release to modulate hippocampal GABAergic transmission. Sci Signal 2019; 12:12/582/eaaw5450. [PMID: 31113852 DOI: 10.1126/scisignal.aaw5450] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Astrocytes are the major glial subtype in the brain and mediate numerous functions ranging from metabolic support to gliotransmitter release through signaling mechanisms controlled by Ca2+ Despite intense interest, the Ca2+ influx pathways in astrocytes remain obscure, hindering mechanistic insights into how Ca2+ signaling is coupled to downstream astrocyte-mediated effector functions. Here, we identified store-operated Ca2+ release-activated Ca2+ (CRAC) channels encoded by Orai1 and STIM1 as a major route of Ca2+ entry for driving sustained and oscillatory Ca2+ signals in astrocytes after stimulation of metabotropic purinergic and protease-activated receptors. Using synaptopHluorin as an optical reporter, we showed that the opening of astrocyte CRAC channels stimulated vesicular exocytosis to mediate the release of gliotransmitters, including ATP. Furthermore, slice electrophysiological recordings showed that activation of astrocytes by protease-activated receptors stimulated interneurons in the CA1 hippocampus to increase inhibitory postsynaptic currents on CA1 pyramidal cells. These results reveal a central role for CRAC channels as regulators of astrocyte Ca2+ signaling, gliotransmitter release, and astrocyte-mediated tonic inhibition of CA1 pyramidal neurons.
Collapse
Affiliation(s)
- Anna B Toth
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kotaro Hori
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michaela M Novakovic
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Natalie G Bernstein
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Laurie Lambot
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
33
|
Yang J, Vitery MDC, Chen J, Osei-Owusu J, Chu J, Qiu Z. Glutamate-Releasing SWELL1 Channel in Astrocytes Modulates Synaptic Transmission and Promotes Brain Damage in Stroke. Neuron 2019; 102:813-827.e6. [PMID: 30982627 DOI: 10.1016/j.neuron.2019.03.029] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 12/14/2018] [Accepted: 03/19/2019] [Indexed: 01/01/2023]
Abstract
By releasing glutamate, astrocytes actively regulate synaptic transmission and contribute to excitotoxicity in neurological diseases. However, the mechanisms of astrocytic glutamate release have been debated. Here, we report non-vesicular release of glutamate through the glutamate-permeable volume-regulated anion channel (VRAC). Both cell swelling and receptor stimulation activated astrocytic VRAC, which requires its only obligatory subunit, Swell1. Astrocyte-specific Swell1 knockout mice exhibited impaired glutamatergic transmission due to the decreases in presynaptic release probability and ambient glutamate level. Consistently, the mutant mice displayed hippocampal-dependent learning and memory deficits. During pathological cell swelling, deletion of astrocytic Swell1 attenuated glutamate-dependent neuronal excitability and protected mice from brain damage after ischemic stroke. Our identification of a new molecular mechanism for channel-mediated glutamate release establishes a role for astrocyte-neuron interactions in both synaptic transmission and brain ischemia. It provides a rationale for targeting VRAC for the treatment of stroke and other neurological diseases associated with excitotoxicity.
Collapse
Affiliation(s)
- Junhua Yang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Maria Del Carmen Vitery
- 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
| | - James Osei-Owusu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jiachen Chu
- Department of Physiology, 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
|
34
|
Gliotransmission: Beyond Black-and-White. J Neurosci 2019; 38:14-25. [PMID: 29298905 DOI: 10.1523/jneurosci.0017-17.2017] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/01/2017] [Accepted: 08/29/2017] [Indexed: 01/09/2023] Open
Abstract
Astrocytes are highly complex cells with many emerging putative roles in brain function. Of these, gliotransmission (active information transfer from glia to neurons) has probably the widest implications on our understanding of how the brain works: do astrocytes really contribute to information processing within the neural circuitry? "Positive evidence" for this stems from work of multiple laboratories reporting many examples of modulatory chemical signaling from astrocytes to neurons in the timeframe of hundreds of milliseconds to several minutes. This signaling involves, but is not limited to, Ca2+-dependent vesicular transmitter release, and results in a variety of regulatory effects at synapses in many circuits that are abolished by preventing Ca2+ elevations or blocking exocytosis selectively in astrocytes. In striking contradiction, methodologically advanced studies by a few laboratories produced "negative evidence," triggering a heated debate on the actual existence and properties of gliotransmission. In this context, a skeptics' camp arose, eager to dismiss the whole positive evidence based on a number of assumptions behind the negative data, such as the following: (1) deleting a single Ca2+ release pathway (IP3R2) removes all the sources for Ca2+-dependent gliotransmission; (2) stimulating a transgenically expressed Gq-GPCR (MrgA1) mimics the physiological Ca2+ signaling underlying gliotransmitter release; (3) age-dependent downregulation of an endogenous GPCR (mGluR5) questions gliotransmitter release in adulthood; and (4) failure by transcriptome analysis to detect vGluts or canonical synaptic SNAREs in astrocytes proves inexistence/functional irrelevance of vesicular gliotransmitter release. We here discuss how the above assumptions are likely wrong and oversimplistic. In light of the most recent literature, we argue that gliotransmission is a more complex phenomenon than originally thought, possibly consisting of multiple forms and signaling processes, whose correct study and understanding require more sophisticated tools and finer scientific experiments than done until today. Under this perspective, the opposing camps can be reconciled and the field moved forward. Along the path, a more cautious mindset and an attitude to open discussion and mutual respect between opponent laboratories will be good companions.Dual Perspectives Companion Paper: Multiple Lines of Evidence Indicate That Gliotransmission Does Not Occur under Physiological Conditions, by Todd A. Fiacco and Ken D. McCarthy.
Collapse
|
35
|
Multiple Lines of Evidence Indicate That Gliotransmission Does Not Occur under Physiological Conditions. J Neurosci 2019; 38:3-13. [PMID: 29298904 DOI: 10.1523/jneurosci.0016-17.2017] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/01/2017] [Accepted: 10/02/2017] [Indexed: 12/22/2022] Open
Abstract
A major controversy persists within the field of glial biology concerning whether or not, under physiological conditions, neuronal activity leads to Ca2+-dependent release of neurotransmitters from astrocytes, a phenomenon known as gliotransmission. Our perspective is that, while we and others can apply techniques to cause gliotransmission, there is considerable evidence gathered using astrocyte-specific and more physiological approaches which suggests that gliotransmission is a pharmacological phenomenon rather than a physiological process. Approaches providing evidence against gliotransmission include stimulation of Gq-GPCRs expressed only in astrocytes, as well as removal of the primary proposed source of astrocyte Ca2+ responsible for gliotransmission. These approaches contrast with those supportive of gliotransmission, which include mechanical stimulation, strong astrocytic depolarization using whole-cell patch-clamp or optogenetics, uncaging Ca2+ or IP3, chelating Ca2+ using BAPTA, and nonspecific bath application of agonists to receptors expressed by a multitude of cell types. These techniques are not subtle and therefore are not supportive of recent suggestions that gliotransmission requires very specific and delicate temporal and spatial requirements. Other evidence, including lack of propagating Ca2+ waves between astrocytes in healthy tissue, lack of expression of vesicular release machinery, and the demise of the d-serine gliotransmission hypothesis, provides additional evidence against gliotransmission. Overall, the data suggest that Ca2+-dependent release of neurotransmitters is the province of neurons, not astrocytes, in the intact brain under physiological conditions.Dual Perspectives Companion Paper: Gliotransmission: Beyond Black-and-White, by Iaroslav Savtchouk and Andrea Volterra.
Collapse
|
36
|
Astrocyte function from information processing to cognition and cognitive impairment. Nat Neurosci 2019; 22:154-166. [DOI: 10.1038/s41593-018-0325-8] [Citation(s) in RCA: 294] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 12/06/2018] [Indexed: 12/22/2022]
|
37
|
|
38
|
Scofield MD. Exploring the Role of Astroglial Glutamate Release and Association With Synapses in Neuronal Function and Behavior. Biol Psychiatry 2018; 84:778-786. [PMID: 29258653 PMCID: PMC5948108 DOI: 10.1016/j.biopsych.2017.10.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/18/2017] [Accepted: 10/31/2017] [Indexed: 12/25/2022]
Abstract
Astrocytes are stellate cells whose appearance can resemble a pointed star, especially when visualizing glial fibrillary acidic protein, a canonical marker for astrocytes. Accordingly, there is a commonly made connection between the points of light that shine in the night sky and the diffuse and abundant cells that buffer ions and provide support for neurons. An exceptional amount of function has been attributed to, negated for, and potentially reaffirmed for these cells, especially regarding their ability to release neuroactive molecules and influence synaptic plasticity. This makes the precise role of astrocytes in tuning neural communication seem difficult to grasp. However, data from animal models of addiction demonstrate that a variety of drug-induced molecular adaptations responsible for relapse vulnerability take place in astrocyte systems that regulate glutamate uptake and release. These findings highlight astrocytes as a critical component of the neural systems responsible for addiction, serving as a key component of the plasticity responsible for relapse and drug seeking. Here I assemble recent findings that utilize genetic tools to selectively manipulate or measure flux of internal calcium in astrocytes, focusing on G protein-coupled receptor-mediated mobilization of calcium and the induction of glutamate release. Further, I compile evidence regarding astrocyte glutamate release as well as astrocyte association with synapses with respect to the impact of these cellular phenomena in shaping synaptic transmission. I also place these findings in the context of the previous studies of Scofield et al., who explored the role of astrocytes in the nucleus accumbens in the neural mechanisms underlying cocaine seeking.
Collapse
Affiliation(s)
- Michael D. Scofield
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, 29425 USA,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425 USA
| |
Collapse
|
39
|
Gómez-Gonzalo M, Zehnder T, Requie LM, Bezzi P, Carmignoto G. Insights into the release mechanism of astrocytic glutamate evoking in neurons NMDA receptor-mediated slow depolarizing inward currents. Glia 2018; 66:2188-2199. [DOI: 10.1002/glia.23473] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/27/2018] [Accepted: 05/25/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Marta Gómez-Gonzalo
- Neuroscience Institute, National Research Council (CNR) and Department of Biomedical Sciences; University of Padova; Padova Italy
| | - Tamara Zehnder
- Department of Fundamental Neurosciences; University of Lausanne; Lausanne Switzerland
| | - Linda Maria Requie
- Neuroscience Institute, National Research Council (CNR) and Department of Biomedical Sciences; University of Padova; Padova Italy
| | - Paola Bezzi
- Department of Fundamental Neurosciences; University of Lausanne; Lausanne Switzerland
| | - Giorgio Carmignoto
- Neuroscience Institute, National Research Council (CNR) and Department of Biomedical Sciences; University of Padova; Padova Italy
| |
Collapse
|
40
|
Wang X, Yu H, You J, Wang C, Feng C, Liu Z, Li Y, Wei R, Xu S, Zhao R, Wu X, Zhang G. Memantine can improve chronic ethanol exposure-induced spatial memory impairment in male C57BL/6 mice by reducing hippocampal apoptosis. Toxicology 2018; 406-407:21-32. [PMID: 29800586 DOI: 10.1016/j.tox.2018.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/17/2018] [Accepted: 05/22/2018] [Indexed: 01/08/2023]
Abstract
Chronic ethanol intake can induce neuronal apoptosis, leading to dementia. We investigated the protective effects of memantine on spatial memory impairment induced by chronic ethanol exposure in mice. Male C57BL/6 mice were administered 10% (m/V) or 20% (m/V) ethanol as the only choice of drinking water. Mice were treated for 60 d, 90 d, or 180 d. Mice were treated with memantine for the same duration (daily 10 mg/kg oral). The Morris water maze and radial arm maze test were used to measure spatial memory. Mice were sacrificed after the behavioral tests. Brains were removed to prepare for paraffin sections, and hippocampi were isolated for protein and RNA extraction. 4',6-diamidino-2-phenylindole (DAPI) staining and immunohistochemical staining of cleaved caspase-3 were performed. Western blot analysis was used to detect the expression of cleaved caspase-3 and calcium-related proteins, including N-methyl-d-aspartic acid receptor 1 (NR1), 1,4,5-trisphosphate receptor 1 (IP3R1), and sarco/endoplasmic reticulum calcium adenosine triphosphatase 1 (SERCA1). The changes of NR1, IP3R1 and SERCA1 mRNA were detected using quantitative polymerase chain reaction (qPCR). The results revealed that chronic ethanol exposure induced spatial memory impairment in mice, as well as increasing the expression of NR1, IP3R1 and SERCA1, the activation of caspase-3 and apoptosis in hippocampus. The effect was particularly prominent in the 20% ethanol group after 180 d exposure. Memantine decreased ethanol-induced spatial memory impairment, caspase-3 activation and apoptosis in the mouse hippocampus. These results suggest that disruption of intracellular calcium balance by ethanol can induce caspase-3 activation and apoptosis, which underlies subsequent spatial memory impairment in mice.
Collapse
Affiliation(s)
- Xiaolong Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Hao Yu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Jiabin You
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Changliang Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Chunmei Feng
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Zhaodi Liu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Ya Li
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Rucheng Wei
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Siqi Xu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Rui Zhao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Xu Wu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China.
| | - Guohua Zhang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China.
| |
Collapse
|
41
|
Mayorquin LC, Rodriguez AV, Sutachan JJ, Albarracín SL. Connexin-Mediated Functional and Metabolic Coupling Between Astrocytes and Neurons. Front Mol Neurosci 2018; 11:118. [PMID: 29695954 PMCID: PMC5905222 DOI: 10.3389/fnmol.2018.00118] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/27/2018] [Indexed: 01/24/2023] Open
Abstract
The central nervous system (CNS) requires sophisticated regulation of neuronal activity. This modulation is partly accomplished by non-neuronal cells, characterized by the presence of transmembrane gap junctions (GJs) and hemichannels (HCs). This allows small molecule diffusion to guarantee neuronal synaptic activity and plasticity. Astrocytes are metabolically and functionally coupled to neurons by the uptake, binding and recycling of neurotransmitters. In addition, astrocytes release metabolites, such as glutamate, glutamine, D-serine, adenosine triphosphate (ATP) and lactate, regulating synaptic activity and plasticity by pre- and postsynaptic mechanisms. Uncoupling neuroglial communication leads to alterations in synaptic transmission that can be detrimental to neuronal circuit function and behavior. Therefore, understanding the pathways and mechanisms involved in this intercellular communication is fundamental for the search of new targets that can be used for several neurological disease treatments. This review will focus on molecular mechanisms mediating physiological and pathological coupling between astrocytes and neurons through GJs and HCs.
Collapse
Affiliation(s)
- Lady C Mayorquin
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Andrea V Rodriguez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Jhon-Jairo Sutachan
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Sonia L Albarracín
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
42
|
Manninen T, Havela R, Linne ML. Computational Models for Calcium-Mediated Astrocyte Functions. Front Comput Neurosci 2018; 12:14. [PMID: 29670517 PMCID: PMC5893839 DOI: 10.3389/fncom.2018.00014] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 02/28/2018] [Indexed: 12/16/2022] Open
Abstract
The computational neuroscience field has heavily concentrated on the modeling of neuronal functions, largely ignoring other brain cells, including one type of glial cell, the astrocytes. Despite the short history of modeling astrocytic functions, we were delighted about the hundreds of models developed so far to study the role of astrocytes, most often in calcium dynamics, synchronization, information transfer, and plasticity in vitro, but also in vascular events, hyperexcitability, and homeostasis. Our goal here is to present the state-of-the-art in computational modeling of astrocytes in order to facilitate better understanding of the functions and dynamics of astrocytes in the brain. Due to the large number of models, we concentrated on a hundred models that include biophysical descriptions for calcium signaling and dynamics in astrocytes. We categorized the models into four groups: single astrocyte models, astrocyte network models, neuron-astrocyte synapse models, and neuron-astrocyte network models to ease their use in future modeling projects. We characterized the models based on which earlier models were used for building the models and which type of biological entities were described in the astrocyte models. Features of the models were compared and contrasted so that similarities and differences were more readily apparent. We discovered that most of the models were basically generated from a small set of previously published models with small variations. However, neither citations to all the previous models with similar core structure nor explanations of what was built on top of the previous models were provided, which made it possible, in some cases, to have the same models published several times without an explicit intention to make new predictions about the roles of astrocytes in brain functions. Furthermore, only a few of the models are available online which makes it difficult to reproduce the simulation results and further develop the models. Thus, we would like to emphasize that only via reproducible research are we able to build better computational models for astrocytes, which truly advance science. Our study is the first to characterize in detail the biophysical and biochemical mechanisms that have been modeled for astrocytes.
Collapse
Affiliation(s)
- Tiina Manninen
- Computational Neuroscience Group, BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
| | | | - Marja-Leena Linne
- Computational Neuroscience Group, BioMediTech Institute and Faculty of Biomedical Sciences and Engineering, Tampere University of Technology, Tampere, Finland
| |
Collapse
|
43
|
Smith NA, Kress BT, Lu Y, Chandler-Militello D, Benraiss A, Nedergaard M. Fluorescent Ca 2+ indicators directly inhibit the Na,K-ATPase and disrupt cellular functions. Sci Signal 2018; 11:11/515/eaal2039. [PMID: 29382785 DOI: 10.1126/scisignal.aal2039] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Fluorescent Ca2+ indicators have been essential for the analysis of Ca2+ signaling events in various cell types. We showed that chemical Ca2+ indicators, but not a genetically encoded Ca2+ indicator, potently suppressed the activity of Na+- and K+-dependent adenosine triphosphatase (Na,K-ATPase), independently of their Ca2+ chelating activity. Loading of commonly used Ca2+ indicators, including Fluo-4 acetoxymethyl (AM), Rhod-2 AM, and Fura-2 AM, and of the Ca2+ chelator BAPTA AM into cultured mouse or human neurons, astrocytes, cardiomyocytes, or kidney proximal tubule epithelial cells suppressed Na,K-ATPase activity by 30 to 80%. Ca2+ indicators also suppressed the agonist-induced activation of the Na,K-ATPase, altered metabolic status, and caused a dose-dependent loss of cell viability. Loading of Ca2+ indicators into mice, which is carried out for two-photon imaging, markedly altered brain extracellular concentrations of K+ and ATP. These results suggest that a critical review of data obtained with chemical Ca2+ indicators may be necessary.
Collapse
Affiliation(s)
- Nathan A Smith
- Center for Translational Neuromedicine, Departments of Neurosurgery and Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA.,Center for Neuroscience Research, Children's Research Institute, Children's National Health System, Washington, DC 20010, USA
| | - Benjamin T Kress
- Center for Translational Neuromedicine, Departments of Neurosurgery and Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yuan Lu
- Center for Translational Neuromedicine, Departments of Neurosurgery and Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Devin Chandler-Militello
- Center for Translational Neuromedicine, Departments of Neurosurgery and Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Abdellatif Benraiss
- Center for Translational Neuromedicine, Departments of Neurosurgery and Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Departments of Neurosurgery and Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
44
|
Rose CR, Felix L, Zeug A, Dietrich D, Reiner A, Henneberger C. Astroglial Glutamate Signaling and Uptake in the Hippocampus. Front Mol Neurosci 2018; 10:451. [PMID: 29386994 PMCID: PMC5776105 DOI: 10.3389/fnmol.2017.00451] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/22/2017] [Indexed: 12/22/2022] Open
Abstract
Astrocytes have long been regarded as essentially unexcitable cells that do not contribute to active signaling and information processing in the brain. Contrary to this classical view, it is now firmly established that astrocytes can specifically respond to glutamate released from neurons. Astrocyte glutamate signaling is initiated upon binding of glutamate to ionotropic and/or metabotropic receptors, which can result in calcium signaling, a major form of glial excitability. Release of so-called gliotransmitters like glutamate, ATP and D-serine from astrocytes in response to activation of glutamate receptors has been demonstrated to modulate various aspects of neuronal function in the hippocampus. In addition to receptors, glutamate binds to high-affinity, sodium-dependent transporters, which results in rapid buffering of synaptically-released glutamate, followed by its removal from the synaptic cleft through uptake into astrocytes. The degree to which astrocytes modulate and control extracellular glutamate levels through glutamate transporters depends on their expression levels and on the ionic driving forces that decrease with ongoing activity. Another major determinant of astrocytic control of glutamate levels could be the precise morphological arrangement of fine perisynaptic processes close to synapses, defining the diffusional distance for glutamate, and the spatial proximity of transporters in relation to the synaptic cleft. In this review, we will present an overview of the mechanisms and physiological role of glutamate-induced ion signaling in astrocytes in the hippocampus as mediated by receptors and transporters. Moreover, we will discuss the relevance of astroglial glutamate uptake for extracellular glutamate homeostasis, focusing on how activity-induced dynamic changes of perisynaptic processes could shape synaptic transmission at glutamatergic synapses.
Collapse
Affiliation(s)
- Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Lisa Felix
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Dirk Dietrich
- Department of Neurosurgery, University of Bonn Medical School, Bonn, Germany
| | - Andreas Reiner
- Cellular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany.,German Center for Degenerative Diseases (DZNE), Bonn, Germany.,Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
45
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
46
|
Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev 2018; 98:239-389. [PMID: 29351512 PMCID: PMC6050349 DOI: 10.1152/physrev.00042.2016] [Citation(s) in RCA: 1076] [Impact Index Per Article: 153.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
47
|
Forsberg D, Ringstedt T, Herlenius E. Astrocytes release prostaglandin E2 to modify respiratory network activity. eLife 2017; 6:29566. [PMID: 28976306 PMCID: PMC5648524 DOI: 10.7554/elife.29566] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/03/2017] [Indexed: 12/31/2022] Open
Abstract
Previously (Forsberg et al., 2016), we revealed that prostaglandin E2 (PGE2), released during hypercapnic challenge, increases calcium oscillations in the chemosensitive parafacial respiratory group (pFRG/RTN). Here, we demonstrate that pFRG/RTN astrocytes are the PGE2 source. Two distinct astrocyte subtypes were found using transgenic mice expressing GFP and MrgA1 receptors in astrocytes. Although most astrocytes appeared dormant during time-lapse calcium imaging, a subgroup displayed persistent, rhythmic oscillating calcium activity. These active astrocytes formed a subnetwork within the respiratory network distinct from the neuronal network. Activation of exogenous MrgA1Rs expressed in astrocytes tripled astrocytic calcium oscillation frequency in both the preBötzinger complex and pFRG/RTN. However, neurons in the preBötC were unaffected, whereas neuronal calcium oscillatory frequency in pFRG/RTN doubled. Notably, astrocyte activation in pFRG/RTN triggered local PGE2 release and blunted the hypercapnic response. Thus, astrocytes play an active role in respiratory rhythm modulation, modifying respiratory-related behavior through PGE2 release in the pFRG/RTN.
Collapse
Affiliation(s)
- David Forsberg
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas Ringstedt
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eric Herlenius
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
48
|
Murphy TR, Davila D, Cuvelier N, Young LR, Lauderdale K, Binder DK, Fiacco TA. Hippocampal and Cortical Pyramidal Neurons Swell in Parallel with Astrocytes during Acute Hypoosmolar Stress. Front Cell Neurosci 2017; 11:275. [PMID: 28979186 PMCID: PMC5611379 DOI: 10.3389/fncel.2017.00275] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/28/2017] [Indexed: 01/08/2023] Open
Abstract
Normal nervous system function is critically dependent on the balance of water and ions in the extracellular space (ECS). Pathological reduction in brain interstitial osmolarity results in osmotically-driven flux of water into cells, causing cellular edema which reduces the ECS and increases neuronal excitability and risk of seizures. Astrocytes are widely considered to be particularly susceptible to cellular edema due to selective expression of the water channel aquaporin-4 (AQP4). The apparent resistance of pyramidal neurons to osmotic swelling has been attributed to lack of functional water channels. In this study we report rapid volume changes in CA1 pyramidal cells in hypoosmolar ACSF (hACSF) that are equivalent to volume changes in astrocytes across a variety of conditions. Astrocyte and neuronal swelling was significant within 1 min of exposure to 17 or 40% hACSF, was rapidly reversible upon return to normosmolar ACSF, and repeatable upon re-exposure to hACSF. Neuronal swelling was not an artifact of patch clamp, occurred deep in tissue, was similar at physiological vs. room temperature, and occurred in both juvenile and adult hippocampal slices. Neuronal swelling was neither inhibited by TTX, nor by antagonists of NMDA or AMPA receptors, suggesting that it was not occurring as a result of excitotoxicity. Surprisingly, genetic deletion of AQP4 did not inhibit, but rather augmented, astrocyte swelling in severe hypoosmolar conditions. Taken together, our results indicate that neurons are not osmoresistant as previously reported, and that osmotic swelling is driven by an AQP4-independent mechanism.
Collapse
Affiliation(s)
- Thomas R. Murphy
- Division of Biomedical Sciences, School of Medicine, University of California, RiversideRiverside, CA, United States
- Center for Glial-Neuronal Interactions, University of California, RiversideRiverside, CA, United States
| | - David Davila
- Center for Glial-Neuronal Interactions, University of California, RiversideRiverside, CA, United States
- Department of Cell Biology and Neuroscience, University of California, RiversideRiverside, CA, United States
| | - Nicholas Cuvelier
- Center for Glial-Neuronal Interactions, University of California, RiversideRiverside, CA, United States
- Department of Cell Biology and Neuroscience, University of California, RiversideRiverside, CA, United States
| | - Leslie R. Young
- Center for Glial-Neuronal Interactions, University of California, RiversideRiverside, CA, United States
- Department of Cell Biology and Neuroscience, University of California, RiversideRiverside, CA, United States
| | - Kelli Lauderdale
- Division of Biomedical Sciences, School of Medicine, University of California, RiversideRiverside, CA, United States
- Center for Glial-Neuronal Interactions, University of California, RiversideRiverside, CA, United States
| | - Devin K. Binder
- Division of Biomedical Sciences, School of Medicine, University of California, RiversideRiverside, CA, United States
- Center for Glial-Neuronal Interactions, University of California, RiversideRiverside, CA, United States
| | - Todd A. Fiacco
- Center for Glial-Neuronal Interactions, University of California, RiversideRiverside, CA, United States
- Department of Cell Biology and Neuroscience, University of California, RiversideRiverside, CA, United States
| |
Collapse
|
49
|
Astrocyte-Mediated Neuronal Synchronization Properties Revealed by False Gliotransmitter Release. J Neurosci 2017; 37:9859-9870. [PMID: 28899919 PMCID: PMC5637115 DOI: 10.1523/jneurosci.2761-16.2017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 06/14/2017] [Accepted: 06/27/2017] [Indexed: 12/21/2022] Open
Abstract
Astrocytes spontaneously release glutamate (Glut) as a gliotransmitter (GT), resulting in the generation of extrasynaptic NMDAR-mediated slow inward currents (SICs) in neighboring neurons, which can increase local neuronal excitability. However, there is a deficit in our knowledge of the factors that control spontaneous astrocyte GT release and the extent of its influence. We found that, in rat brain slices, increasing the supply of the physiological transmitter Glut increased the frequency and signaling charge of SICs over an extended period. This phenomenon was replicated by exogenous preexposure to the amino acid D-aspartate (D-Asp). Using D-Asp as a “false” GT, we determined the extent of local neuron excitation by GT release in ventrobasal thalamus, CA1 hippocampus, and somatosensory cortex. By analyzing synchronized neuronal NMDAR-mediated excitation, we found that the properties of the excitation were conserved in different brain areas. In the three areas, astrocyte-derived GT release synchronized groups of neurons at distances of >;200 μm. Individual neurons participated in more than one synchronized population, indicating that individual neurons can be excited by more than one astrocyte and that individual astrocytes may determine a neuron's synchronized network. The results confirm that astrocytes can act as excitatory nodes that can influence neurons over a significant range in a number of brain regions. Our findings further suggest that chronic elevation of ambient Glut levels can lead to increased GT Glut release, which may be relevant in some pathological states. SIGNIFICANCE STATEMENT Astrocytes spontaneously release glutamate (Glut) and other gliotransmitters (GTs) that can modify neuronal activity. Exposing brain slices to Glut and D-aspartate (D-Asp) before recording resulted in an increase in frequency of GT-mediated astrocyte–neuron signaling. Using D-Asp, it was possible to investigate the effects of specific GT release at neuronal NMDARs. Calcium imaging showed synchronized activity in groups of neurons in cortex, hippocampus, and thalamus. The size of these populations was similar in all areas and some neurons were involved in more than one synchronous group. The findings show that GT release is supply dependent and that the properties of the signaling and activated networks are largely conserved between different brain areas.
Collapse
|
50
|
Contribution of Astroglial Cx43 Hemichannels to the Modulation of Glutamatergic Currents by D-Serine in the Mouse Prefrontal Cortex. J Neurosci 2017; 37:9064-9075. [PMID: 28821660 DOI: 10.1523/jneurosci.2204-16.2017] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 05/16/2017] [Accepted: 06/10/2017] [Indexed: 11/21/2022] Open
Abstract
Astrocytes interact dynamically with neurons by modifying synaptic activity and plasticity. This interplay occurs through a process named gliotransmission, meaning that neuroactive molecules are released by astrocytes. Acting as a gliotransmitter, D-serine, a co-agonist of the NMDA receptor at the glycine-binding site, can be released by astrocytes in a calcium [Ca2+]i-dependent manner. A typical feature of astrocytes is their high expression level of connexin43 (Cx43), a protein forming gap junction channels and hemichannels associated with dynamic neuroglial interactions. Pharmacological and genetic inhibition of Cx43 hemichannel activity reduced the amplitude of NMDA EPSCs in mouse layer 5 prefrontal cortex pyramidal neurons without affecting AMPA EPSC currents. This reduction of NMDA EPSCs was rescued by addition of D-serine in the extracellular medium. LTP of NMDA and AMPA EPSCs after high-frequency stimulation was reduced by prior inhibition of Cx43 hemichannel activity. Inactivation of D-serine synthesis within the astroglial network resulted in the reduction of NMDA EPSCs, which was rescued by adding extracellular D-serine. We showed that the activity of Cx43 hemichannels recorded in cultured astrocytes was [Ca2+]I dependent. Accordingly, in acute cortical slices, clamping [Ca2+]i at a low level in astroglial network resulted in an inhibition of NMDA EPSC potentiation that was rescued by adding extracellular D-serine. This work demonstrates that astroglial Cx43 hemichannel activity is associated with D-serine release. This process, occurring by direct permeation of D-serine through hemichannels or indirectly by Ca2+ entry and activation of other [Ca2+]i-dependent mechanisms results in the modulation of synaptic activity and plasticity.SIGNIFICANCE STATEMENT We recorded neuronal glutamatergic (NMDA and AMPA) responses in prefrontal cortex (PFC) neurons and used pharmacological and genetic interventions to block connexin-mediated hemichannel activity specifically in a glial cell population. For the first time in astrocytes, we demonstrated that hemichannel activity depends on the intracellular calcium concentration and is associated with D-serine release. Blocking hemichannel activity reduced the LTP of these excitatory synaptic currents triggered by high-frequency stimulation. These observations may be particularly relevant in the PFC, where D-serine and its converting enzyme are highly expressed.
Collapse
|