1
|
Wang Z, Wei X, Duan L. Regulatory mechanism of inhibitory interneurons with time-delay on epileptic seizures under sinusoidal sensory stimulation. Cogn Neurodyn 2025; 19:37. [PMID: 39926336 PMCID: PMC11799515 DOI: 10.1007/s11571-025-10227-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/12/2025] [Accepted: 01/23/2025] [Indexed: 02/11/2025] Open
Abstract
Epilepsy is a neurological disorder in which complex electrophysiological processes are closely linked to inherent nonlinear kinetic properties. This study investigates the effects of sinusoidal sensory stimulation bias and time-delay on the dynamics of epileptic seizures within a corticothalamic neural network model. The results indicate that an increase in sensory stimulation bias can prematurely terminate seizures, and high-frequency stimulation can induce a phenomenon of frequency resonance. Meanwhile, discharge states transitions are associated with the emergence of bifurcation points. Time-delay exerts a significant regulatory influence on pathways with delay embedding (I2-PY), whereas its impact on pathways without delay embedding (I1-I1 and thalamic relay nucleus (TC)-I2) is negligible. Under sinusoidal sensory stimulation, the responses of three pathways (I1-I1, I1-PY, and I2-PY) associated with inhibitory interneurons reveal that the inhibitory properties of interneurons can suppress seizures; however, an excessively strong inhibitory effect may also precipitate seizures and facilitate state transitions. These findings contribute to a deeper understanding of seizure dynamics and may guide future research in the transmission and evolution of seizures.
Collapse
Affiliation(s)
- Zhihui Wang
- College of Science, North China University of Technology, Beijing, 100144 China
| | - Xindan Wei
- College of Science, North China University of Technology, Beijing, 100144 China
| | - Lixia Duan
- College of Science, North China University of Technology, Beijing, 100144 China
| |
Collapse
|
2
|
Guan L, Qiu M, Li N, Zhou Z, Ye R, Zhong L, Xu Y, Ren J, Liang Y, Shao X, Fang J, Fang J, Du J. Inhibitory gamma-aminobutyric acidergic neurons in the anterior cingulate cortex participate in the comorbidity of pain and emotion. Neural Regen Res 2025; 20:2838-2854. [PMID: 39314159 PMCID: PMC11826466 DOI: 10.4103/nrr.nrr-d-24-00429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/19/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Pain is often comorbid with emotional disorders such as anxiety and depression. Hyperexcitability of the anterior cingulate cortex has been implicated in pain and pain-related negative emotions that arise from impairments in inhibitory gamma-aminobutyric acid neurotransmission. This review primarily aims to outline the main circuitry (including the input and output connectivity) of the anterior cingulate cortex and classification and functions of different gamma-aminobutyric acidergic neurons; it also describes the neurotransmitters/neuromodulators affecting these neurons, their intercommunication with other neurons, and their importance in mental comorbidities associated with chronic pain disorders. Improving understanding on their role in pain-related mental comorbidities may facilitate the development of more effective treatments for these conditions. However, the mechanisms that regulate gamma-aminobutyric acidergic systems remain elusive. It is also unclear as to whether the mechanisms are presynaptic or postsynaptic. Further exploration of the complexities of this system may reveal new pathways for research and drug development.
Collapse
Affiliation(s)
- Lu Guan
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Mengting Qiu
- Fuchun Community Health Service Center of Fuyang District, Hangzhou, Zhejiang Province, China
| | - Na Li
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhengxiang Zhou
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Ru Ye
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Liyan Zhong
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yashuang Xu
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Junhui Ren
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi Liang
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaomei Shao
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jianqiao Fang
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Junfan Fang
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Junying Du
- Department of Neurobiology and Acupuncture Research, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- Key Laboratory for Research of Acupuncture Treatment and Transformation of Emotional Diseases, Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
3
|
Alberquilla S, Nanclares C, Expósito S, Gall G, Kofuji P, Araque A, Martín ED, Moratalla R. Astrocytes Mediate Psychostimulant-Induced Alterations of Spike-Timing Dependent Synaptic Plasticity. Glia 2025; 73:1051-1067. [PMID: 39801264 PMCID: PMC11920680 DOI: 10.1002/glia.24672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 03/20/2025]
Abstract
At cellular and circuit levels, drug addiction is considered a dysregulation of synaptic plasticity. In addition, dysfunction of the glutamate transporter 1 (GLT-1) in the nucleus accumbens (NAc) has also been proposed as a mechanism underlying drug addiction. However, the cellular and synaptic impact of GLT-1 alterations in the NAc remain unclear. Here we show in the NAc that 10 days withdraw after 5 days treatment with cocaine or amphetamine decreases GLT-1 expression in astrocytes, which results in the prolongation of the excitatory postsynaptic potential (EPSP) decay kinetics in D1 receptor-containing medium spiny neurons (D1R-MSNs). Using the spike timing dependent plasticity (STDP) paradigm, we found that enlargement of EPSP duration results in switching the LTP elicited in control animals to LTD in psychostimulant-treated mice. In contrast to D1-MSNs, D2-MSNs did not display changes in EPSP kinetics and synaptic plasticity. Notably, the psychostimulant-induced synaptic transmission and synaptic plasticity effects were absent in IP3R2-/- mice, which lack astrocyte calcium signal, but were mimicked by the selective astrocytes stimulation with DREADDs. Finally, ceftriaxone, which upregulates GLT-1, restored normal GLT-1 function, EPSP kinetics, and synaptic plasticity in psychostimulant-treated mice. Therefore, we propose that cocaine and amphetamine increase dopaminergic levels in the NAc, which stimulates astrocytes and downregulates the GLT-1. The decreased GLT-1 function prolonged the EPSP kinetics, leading to the modulation of the STDP, transforming the LTP observed in control animals into LTD in psychostimulant-treated mice. Present work reveals a novel mechanism underlying the synaptic plasticity changes induced by these drugs of abuse.
Collapse
Affiliation(s)
- Samuel Alberquilla
- Cajal Institute, CSICMadridSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
| | - Carmen Nanclares
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Grace Gall
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Paulo Kofuji
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Alfonso Araque
- Department of NeuroscienceUniversity of MinnesotaMinneapolisMinnesotaUSA
| | | | - Rosario Moratalla
- Cajal Institute, CSICMadridSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Ammothumkandy A, Cayce A, Shariq M, Bonaguidi MA. Astroglia's role in synchronized spontaneous neuronal activity: from physiology to pathology. Front Cell Neurosci 2025; 19:1544460. [PMID: 40177583 PMCID: PMC11961896 DOI: 10.3389/fncel.2025.1544460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
The nervous system relies on a balance of excitatory and inhibitory signals. Aberrant neuronal hyperactivity is a pathological phenotype associated with several neurological disorders, with its most severe effects observed in epilepsy patients. This review explores the literature on spontaneous synchronized neuronal activity, its physiological role, and its aberrant forms in disease. Emphasizing the importance of targeting underlying disease mechanisms beyond traditional neuron-focused therapies, the review delves into the role of astroglia in epilepsy progression. We detail how astroglia transitions from a normal to a pathological state, leading to epileptogenic seizures and cognitive decline. Astroglia activity is correlated with epileptiform activity in both animal models and human tissue, indicating their potential role in seizure induction and modulation. Understanding astroglia's dual beneficial and detrimental roles could lead to novel treatments for epilepsy and other neurological disorders with aberrant neuronal activity as the underlying disease substrate.
Collapse
Affiliation(s)
- Aswathy Ammothumkandy
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Alisha Cayce
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Mohammad Shariq
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Michael A. Bonaguidi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
- Keck School of Medicine, Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
- Department of Gerontology, University of Southern California, Los Angeles, CA, United States
- Keck School of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
7
|
Gómez-Gonzalo M. Astrocytes in Rodent Anxiety-Related Behavior: Role of Calcium and Beyond. Int J Mol Sci 2025; 26:2774. [PMID: 40141416 PMCID: PMC11943343 DOI: 10.3390/ijms26062774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Anxiety is a physiological, emotional response that anticipates distal threats. When kept under control, anxiety is a beneficial response, helping animals to maintain heightened attention in environments with potential dangers. However, an overestimation of potential threats can lead to an excessive expression of anxiety that, in humans, may evolve into anxiety disorders. Pharmacological treatments show variable efficacy among patients, highlighting the need for more efforts to better understand the pathogenesis of anxiety disorders. Mounting evidence suggests that astrocytes, a type of glial cells, are active partners of neurons in brain circuits and in the regulation of behaviors under both physiological and pathological conditions. In this review, I summarize the current literature on the role of astrocytes from different brain regions in modulating anxious states, with the goal of exploring novel cerebral mechanisms to identify potential innovative therapeutic targets for the treatment of anxiety disorders.
Collapse
Affiliation(s)
- Marta Gómez-Gonzalo
- Section of Padua, Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| |
Collapse
|
8
|
Kékesi O, Keembiyage N, Buskila Y. Calcium Imaging in Brain Tissue Slices. Methods Mol Biol 2025; 2861:89-96. [PMID: 39395099 DOI: 10.1007/978-1-0716-4164-4_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Calcium imaging is a method that was first developed in the mid-1970s yet kept developing until current days to allow accurate measurement of free calcium ions in tissues. This widely used method has provided significant advances to our understanding of cellular signal transduction, including the discovery of subcellular compartmentalization of neurons and astrocytes, the identification of multiple signaling pathways, and mapping the functional connectivity between astrocytes and neuronal networks. Here we describe a method for the loading and imaging of cell-permeable AM ester calcium-sensitive dyes for the in vitro measurement of free intracellular Ca2+ ions in acute brain slices.
Collapse
Affiliation(s)
- Orsolya Kékesi
- School of Biomedical Engineering, The University of Sydney, Camperdown, NSW, Australia.
| | - Nisal Keembiyage
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- The MARCS Institute, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
9
|
Dzyubenko E, Hermann DM. Neuroglia and extracellular matrix molecules. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:197-211. [PMID: 40122625 DOI: 10.1016/b978-0-443-19104-6.00010-3] [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 provides a comprehensive overview of the roles of astrocytes, microglia, and the extracellular matrix (ECM) in regulating neuroplasticity and maintaining brain homeostasis. Astrocytes provide essential metabolic support to neurons, regulate synapse development, support neuroplasticity mechanisms, and modulate neurotransmission. Microglia, the resident immune cells of the brain, play a critical role in neuroinflammatory responses and homeostatic brain regulation by modulating synapse formation and pruning. The extracellular space (ECS) mediates intercellular interactions, provides a highly regulated environment for intercellular communication, and is filled with ECM molecules. Proteoglycans and polysaccharides of the ECM play a vital role not only in brain development but also in brain function throughout life. In the injured brain, neuroplasticity and regeneration can be bidirectionally regulated as a result of the interplay between ECM, astrocytes, and microglia. The modulation of synaptic strength, structural remodeling, and modification of intrinsic neuronal properties are among the central mechanisms that contribute to neuronal plasticity in health and disease. We believe that the understanding of ECM-glia interactions and their role in neuroplasticity regulation is key to the development of novel therapeutic strategies in neurologic disorders.
Collapse
Affiliation(s)
- Egor Dzyubenko
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Dirk M Hermann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
10
|
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
|
11
|
Lines J, Baraibar A, Nanclares C, Martin ED, Aguilar J, Kofuji P, Navarrete M, Araque A. A spatial threshold for astrocyte calcium surge. eLife 2024; 12:RP90046. [PMID: 39680037 DOI: 10.7554/elife.90046] [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: 12/17/2024] Open
Abstract
Astrocytes are active cells involved in brain function through the bidirectional communication with neurons, in which astrocyte calcium plays a crucial role. Synaptically evoked calcium increases can be localized to independent subcellular domains or expand to the entire cell, i.e., calcium surge. Because a single astrocyte may contact ~100,000 synapses, the control of the intracellular calcium signal propagation may have relevant consequences on brain function. Yet, the properties governing the spatial dynamics of astrocyte calcium remains poorly defined. Imaging subcellular responses of cortical astrocytes to sensory stimulation in mice, we show that sensory-evoked astrocyte calcium responses originated and remained localized in domains of the astrocytic arborization, but eventually propagated to the entire cell if a spatial threshold of >23% of the arborization being activated was surpassed. Using Itpr2-/- mice, we found that type-2 IP3 receptors were necessary for the generation of astrocyte calcium surge. We finally show using in situ electrophysiological recordings that the spatial threshold of the astrocyte calcium signal consequently determined the gliotransmitter release. Present results reveal a fundamental property of astrocyte physiology, i.e., a spatial threshold for astrocyte calcium propagation, which depends on astrocyte intrinsic properties and governs astrocyte integration of local synaptic activity and subsequent neuromodulation.
Collapse
Affiliation(s)
- Justin Lines
- Department of Neuroscience, University of Minnesota, Minneapolis, United States
| | - Andres Baraibar
- Department of Neuroscience, University of Minnesota, Minneapolis, United States
| | - Carmen Nanclares
- Department of Neuroscience, University of Minnesota, Minneapolis, United States
| | | | - Juan Aguilar
- Experimental Neurophysiology Group, Hospital Nacional de Parapléjicos SESCAM, Toledo, Spain
| | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, United States
| | | | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, United States
| |
Collapse
|
12
|
Kosenkov AM, Mal'tseva VN, Maiorov SA, Gaidin SG. The role of the endocannabinoid system in the pathogenesis and treatment of epilepsy. Rev Neurosci 2024:revneuro-2024-0114. [PMID: 39660979 DOI: 10.1515/revneuro-2024-0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024]
Abstract
Epilepsy is a group of chronic neurological brain disorders characterized by recurrent spontaneous unprovoked seizures, which are accompanied by significant neurobiological, cognitive, and psychosocial impairments. With a global prevalence of approximately 0.5-1 % of the population, epilepsy remains a serious public health concern. Despite the development and widespread use of over 20 anticonvulsant drugs, around 30 % of patients continue to experience drug-resistant seizures, leading to a substantial reduction in quality of life and increased mortality risk. Given the limited efficacy of current treatments, exploring new therapeutic approaches is critically important. In recent years, Gi-protein-coupled receptors, particularly cannabinoid receptors CB1 and CB2, have garnered increasing attention as promising targets for the treatment seizures and prevention of epilepsy. Emerging evidence suggests a significant role of the cannabinoid system in modulating neuronal activity and protecting against hyperexcitability, underscoring the importance of further research in this area. This review provides up-to-date insights into the pathogenesis and treatment of epilepsy, with a special focus on the role of the cannabinoid system, highlighting the need for continued investigation to develop more effective therapeutic strategies.
Collapse
Affiliation(s)
- Artem M Kosenkov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russian Federation
| | - Valentina N Mal'tseva
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russian Federation
| | - Sergei A Maiorov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russian Federation
| | - Sergei G Gaidin
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russian Federation
| |
Collapse
|
13
|
Cases‐Cunillera S, Quatraccioni A, Rossini L, Ruffolo G, Ono T, Baulac S, Auvin S, O'Brien TJ, Henshall DC, Akman Ö, Sankar R, Galanopoulou AS. WONOEP appraisal: The role of glial cells in focal malformations associated with early onset epilepsies. Epilepsia 2024; 65:3457-3468. [PMID: 39401070 PMCID: PMC11647439 DOI: 10.1111/epi.18126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024]
Abstract
Epilepsy represents a common neurological disorder in patients with developmental brain lesions, particularly in association with malformations of cortical development and low-grade glioneuronal tumors. In these diseases, genetic and molecular alterations in neurons are increasingly discovered that can trigger abnormalities in the neuronal network, leading to higher neuronal excitability levels. However, the mechanisms underlying epilepsy cannot rely solely on assessing the neuronal component. Growing evidence has revealed the high degree of complexity underlying epileptogenic processes, in which glial cells emerge as potential modulators of neuronal activity. Understanding the role of glial cells in developmental brain lesions such as malformations of cortical development and low-grade glioneuronal tumors is crucial due to the high degree of pharmacoresistance characteristic of these lesions. This has prompted research to investigate the role of glial and immune cells in epileptiform activity to find new therapeutic targets that could be used as combinatorial drug therapy. In a special session of the XVI Workshop of the Neurobiology of Epilepsy (WONOEP, Talloires, France, July 2022) organized by the Neurobiology Commission of the International League Against Epilepsy, we discussed the evidence exploring the genetic and molecular mechanisms of glial cells and immune response and their implications in the pathogenesis of neurodevelopmental pathologies associated with early life epilepsies.
Collapse
Affiliation(s)
- Silvia Cases‐Cunillera
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Neuronal Signaling in Epilepsy and GliomaParisFrance
| | - Anne Quatraccioni
- Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of BonnBonnGermany
| | - Laura Rossini
- Epilepsy UnitFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Gabriele Ruffolo
- Department of Physiology and PharmacologyIstituto Pasteur–Fondazione Cenci Bolognetti, University of Rome SapienzaRomeItaly
- IRCCS San Raffaele RomaRomeItaly
| | - Tomonori Ono
- Epilepsy Center, National Hospital Organization Nagasaki Medical CenterŌmuraJapan
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau–Paris Brain Institute–ICM, INSERM, CNRS, AP‐HP, Hôpital de la Pitié SalpêtrièreParisFrance
| | - Stéphane Auvin
- Pediatric Neurology Department, AP‐HP, Robert Debré University HospitalCRMR épilepsies Rares, EpiCARE memberParisFrance
- Université Paris Cité, INSERM NeuroDiderotParisFrance
- Institut Universitaire de FranceParisFrance
| | - Terence J. O'Brien
- Department of Neuroscience, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of NeurologyRoyal Melbourne HospitalMelbourneVictoriaAustralia
- Department of NeurologyAlfred HealthMelbourneVictoriaAustralia
- Department of Medicine (Royal Melbourne Hospital)University of MelbourneMelbourneVictoriaAustralia
| | - David C. Henshall
- Department of Physiology and Medical Physics, RCSIUniversity of Medicine and Health SciencesDublinIreland
| | - Özlem Akman
- Department of PhysiologyFaculty of Medicine, Demiroglu Bilim UniversityIstanbulTurkey
| | - Raman Sankar
- Department of Pediatrics and NeurologyDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Aristea S. Galanopoulou
- Saul R. Korey Department of Neurology, Isabelle Rapin Division of Child Neurology, Dominique P. Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
14
|
Park J, Sipe GO, Tang X, Ojha P, Fernandes G, Leow YN, Zhang C, Osako Y, Natesan A, Drummond GT, Jaenisch R, Sur M. Astrocytic modulation of population encoding in mouse visual cortex via GABA transporter 3 revealed by multiplexed CRISPR/Cas9 gene editing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622321. [PMID: 39605420 PMCID: PMC11601534 DOI: 10.1101/2024.11.06.622321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Astrocytes, which are increasingly recognized as pivotal constituents of brain circuits governing a wide range of functions, express GABA transporter 3 (Gat3), an astrocyte-specific GABA transporter responsible for maintenance of extra-synaptic GABA levels. Here, we examined the functional role of Gat3 in astrocyte-mediated modulation of neuronal activity and information encoding. First, we developed a multiplexed CRISPR construct applicable for effective genetic ablation of Gat3 in the visual cortex of adult mice. Using in vivo two-photon calcium imaging of visual cortex neurons in Gat3 knockout mice, we observed changes in spontaneous and visually driven single neuronal response properties such as response magnitudes and trial-to-trial variability. Gat3 knockout exerted a pronounced influence on population-level neuronal activity, altering the response dynamics of neuronal populations and impairing their ability to accurately represent stimulus information. These findings demonstrate that Gat3 in astrocytes profoundly shapes the sensory information encoding capacity of neurons and networks within the visual cortex.
Collapse
Affiliation(s)
- Jiho Park
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- These authors contributed equally
| | - Grayson O. Sipe
- Department of Biology, Eberly College of Science and Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
- These authors contributed equally
| | - Xin Tang
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA, 02115, USA
| | - Prachi Ojha
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Giselle Fernandes
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Yi Ning Leow
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Caroline Zhang
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
| | - Yuma Osako
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Arundhati Natesan
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Gabrielle T. Drummond
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA, 02142, USA
| | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 01239, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
15
|
Sanz-Gálvez R, Falardeau D, Kolta A, Inglebert Y. The role of astrocytes from synaptic to non-synaptic plasticity. Front Cell Neurosci 2024; 18:1477985. [PMID: 39493508 PMCID: PMC11527691 DOI: 10.3389/fncel.2024.1477985] [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: 08/08/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024] Open
Abstract
Information storage and transfer in the brain require a high computational power. Neuronal network display various local or global mechanisms to allow information storage and transfer in the brain. From synaptic to intrinsic plasticity, the rules of input-output function modulation have been well characterized in neurons. In the past years, astrocytes have been suggested to increase the computational power of the brain and we are only just starting to uncover their role in information processing. Astrocytes maintain a close bidirectional communication with neurons to modify neuronal network excitability, transmission, axonal conduction, and plasticity through various mechanisms including the release of gliotransmitters or local ion homeostasis. Astrocytes have been significantly studied in the context of long-term or short-term synaptic plasticity, but this is not the only mechanism involved in memory formation. Plasticity of intrinsic neuronal excitability also participates in memory storage through regulation of voltage-gated ion channels or axonal morphological changes. Yet, the contribution of astrocytes to these other forms of non-synaptic plasticity remains to be investigated. In this review, we summarized the recent advances on the role of astrocytes in different forms of plasticity and discuss new directions and ideas to be explored regarding astrocytes-neuronal communication and regulation of plasticity.
Collapse
Affiliation(s)
- Rafael Sanz-Gálvez
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage (CIRCA), Montréal, QC, Canada
| | - Dominic Falardeau
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage (CIRCA), Montréal, QC, Canada
| | - Arlette Kolta
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage (CIRCA), Montréal, QC, Canada
- Department of Stomatology, Université de Montréal, Montréal, QC, Canada
| | - Yanis Inglebert
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage (CIRCA), Montréal, QC, Canada
| |
Collapse
|
16
|
Xie L, Wu Q, Li K, Khan MAS, Zhang A, Sinha B, Li S, Chang SL, Brody DL, Grinstaff MW, Zhou S, Alterovitz G, Liu P, Wang X. Tryptophan Metabolism in Alzheimer's Disease with the Involvement of Microglia and Astrocyte Crosstalk and Gut-Brain Axis. Aging Dis 2024; 15:2168-2190. [PMID: 38916729 PMCID: PMC11346405 DOI: 10.14336/ad.2024.0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/03/2024] [Indexed: 06/26/2024] Open
Abstract
Alzheimer's disease (AD) is an age-dependent neurodegenerative disease characterized by extracellular Amyloid Aβ peptide (Aβ) deposition and intracellular Tau protein aggregation. Glia, especially microglia and astrocytes are core participants during the progression of AD and these cells are the mediators of Aβ clearance and degradation. The microbiota-gut-brain axis (MGBA) is a complex interactive network between the gut and brain involved in neurodegeneration. MGBA affects the function of glia in the central nervous system (CNS), and microbial metabolites regulate the communication between astrocytes and microglia; however, whether such communication is part of AD pathophysiology remains unknown. One of the potential links in bilateral gut-brain communication is tryptophan (Trp) metabolism. The microbiota-originated Trp and its metabolites enter the CNS to control microglial activation, and the activated microglia subsequently affect astrocyte functions. The present review highlights the role of MGBA in AD pathology, especially the roles of Trp per se and its metabolism as a part of the gut microbiota and brain communications. We (i) discuss the roles of Trp derivatives in microglia-astrocyte crosstalk from a bioinformatics perspective, (ii) describe the role of glia polarization in the microglia-astrocyte crosstalk and AD pathology, and (iii) summarize the potential of Trp metabolism as a therapeutic target. Finally, we review the role of Trp in AD from the perspective of the gut-brain axis and microglia, as well as astrocyte crosstalk, to inspire the discovery of novel AD therapeutics.
Collapse
Affiliation(s)
- Lushuang Xie
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Qiaofeng Wu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Kelin Li
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Department of Chemistry, Boston University, Boston, MA 02215, USA.
| | - Mohammed A. S. Khan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Andrew Zhang
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Bharati Sinha
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Sihui Li
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Sulie L. Chang
- Department of Biological Sciences, Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ 07079, USA.
| | - David L. Brody
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | | | - Shuanhu Zhou
- Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02115, USA.
| | - Gil Alterovitz
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Pinghua Liu
- Department of Chemistry, Boston University, Boston, MA 02215, USA.
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Lines J, Baraibar A, Nanclares C, Martín ED, Aguilar J, Kofuji P, Navarrete M, Araque A. A spatial threshold for astrocyte calcium surge. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.18.549563. [PMID: 37503130 PMCID: PMC10370153 DOI: 10.1101/2023.07.18.549563] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Astrocytes are active cells involved in brain function through the bidirectional communication with neurons, in which the astrocyte calcium signal plays a crucial role. Synaptically-evoked calcium increases can be localized to independent subcellular domains or expand to the entire cell, i.e., calcium surge. In turn, astrocytes may regulate individual synapses by calcium-dependent release of gliotransmitters. Because a single astrocyte may contact ∼100,000 synapses, the control of the intracellular calcium signal propagation may have relevant consequences on brain function by regulating the spatial range of astrocyte neuromodulation of synapses. Yet, the properties governing the spatial dynamics of the astrocyte calcium signal remains poorly defined. Imaging subcellular responses of cortical astrocytes to sensory stimulation in mice, we show that sensory-evoked astrocyte calcium responses originated and remained localized in domains of the astrocytic arborization, but eventually propagated to the entire cell if a spatial threshold of >23% of the arborization being activated was surpassed. Using transgenic IP 3 R2 -/- mice, we found that type-2 IP 3 receptors were necessary for the generation of the astrocyte calcium surge. We finally show using in situ electrophysiological recordings that the spatial threshold of the astrocyte calcium signal consequently determined the gliotransmitter release. Present results reveal a fundamental property of astrocyte calcium physiology, i.e., a spatial threshold for the astrocyte intracellular calcium signal propagation, which depends on astrocyte intrinsic properties and governs the astrocyte integration of local synaptic activity and the subsequent neuromodulation. One-Sentence Summary There is a spatial threshold for the astrocyte intracellular calcium signal propagation that is determined by astrocyte intrinsic properties and controls gliotransmission.
Collapse
|
18
|
Oliinyk D, Will A, Schneidmadel FR, Böhme M, Rinke J, Hochhaus A, Ernst T, Hahn N, Geis C, Lubeck M, Raether O, Humphrey SJ, Meier F. µPhos: a scalable and sensitive platform for high-dimensional phosphoproteomics. Mol Syst Biol 2024; 20:972-995. [PMID: 38907068 PMCID: PMC11297287 DOI: 10.1038/s44320-024-00050-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
Mass spectrometry has revolutionized cell signaling research by vastly simplifying the analysis of many thousands of phosphorylation sites in the human proteome. Defining the cellular response to perturbations is crucial for further illuminating the functionality of the phosphoproteome. Here we describe µPhos ('microPhos'), an accessible phosphoproteomics platform that permits phosphopeptide enrichment from 96-well cell culture and small tissue amounts in <8 h total processing time. By greatly minimizing transfer steps and liquid volumes, we demonstrate increased sensitivity, >90% selectivity, and excellent quantitative reproducibility. Employing highly sensitive trapped ion mobility mass spectrometry, we quantify ~17,000 Class I phosphosites in a human cancer cell line using 20 µg starting material, and confidently localize ~6200 phosphosites from 1 µg. This depth covers key signaling pathways, rendering sample-limited applications and perturbation experiments with hundreds of samples viable. We employ µPhos to study drug- and time-dependent response signatures in a leukemia cell line, and by quantifying 30,000 Class I phosphosites in the mouse brain we reveal distinct spatial kinase activities in subregions of the hippocampal formation.
Collapse
Affiliation(s)
- Denys Oliinyk
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
| | - Andreas Will
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
| | - Felix R Schneidmadel
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
| | - Maximilian Böhme
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Jenny Rinke
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Andreas Hochhaus
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Thomas Ernst
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Nina Hahn
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Christian Geis
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Markus Lubeck
- Bruker Daltonics GmbH & Co. KG, 28359, Bremen, Germany
| | | | - Sean J Humphrey
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052, Victoria, Australia.
| | - Florian Meier
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany.
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany.
| |
Collapse
|
19
|
Frago LM, Gómez-Romero A, Collado-Pérez R, Argente J, Chowen JA. Synergism Between Hypothalamic Astrocytes and Neurons in Metabolic Control. Physiology (Bethesda) 2024; 39:0. [PMID: 38530221 DOI: 10.1152/physiol.00009.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/05/2024] [Accepted: 03/22/2024] [Indexed: 03/27/2024] Open
Abstract
Astrocytes are no longer considered as passive support cells. In the hypothalamus, these glial cells actively participate in the control of appetite, energy expenditure, and the processes leading to obesity and its secondary complications. Here we briefly review studies supporting this conclusion and the advances made in understanding the underlying mechanisms.
Collapse
Affiliation(s)
- Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Gómez-Romero
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, Campus of International Excellence, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, Campus of International Excellence, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
20
|
Birgül Iyison N, Abboud C, Abboud D, Abdulrahman AO, Bondar AN, Dam J, Georgoussi Z, Giraldo J, Horvat A, Karoussiotis C, Paz-Castro A, Scarpa M, Schihada H, Scholz N, Güvenc Tuna B, Vardjan N. ERNEST COST action overview on the (patho)physiology of GPCRs and orphan GPCRs in the nervous system. Br J Pharmacol 2024. [PMID: 38825750 DOI: 10.1111/bph.16389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/09/2024] [Accepted: 02/24/2024] [Indexed: 06/04/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of cell surface receptors that play a critical role in nervous system function by transmitting signals between cells and their environment. They are involved in many, if not all, nervous system processes, and their dysfunction has been linked to various neurological disorders representing important drug targets. This overview emphasises the GPCRs of the nervous system, which are the research focus of the members of ERNEST COST action (CA18133) working group 'Biological roles of signal transduction'. First, the (patho)physiological role of the nervous system GPCRs in the modulation of synapse function is discussed. We then debate the (patho)physiology and pharmacology of opioid, acetylcholine, chemokine, melatonin and adhesion GPCRs in the nervous system. Finally, we address the orphan GPCRs, their implication in the nervous system function and disease, and the challenges that need to be addressed to deorphanize them.
Collapse
Affiliation(s)
- Necla Birgül Iyison
- Department of Molecular Biology and Genetics, University of Bogazici, Istanbul, Turkey
| | - Clauda Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | - Dayana Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liege, Liege, Belgium
| | | | - Ana-Nicoleta Bondar
- Faculty of Physics, University of Bucharest, Magurele, Romania
- Forschungszentrum Jülich, Institute for Computational Biomedicine (IAS-5/INM-9), Jülich, Germany
| | - Julie Dam
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
| | - Zafiroula Georgoussi
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Madrid, Spain
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anemari Horvat
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Christos Karoussiotis
- Laboratory of Cellular Signalling and Molecular Pharmacology, Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Alba Paz-Castro
- Molecular Pharmacology of GPCRs research group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago, Spain
| | - Miriam Scarpa
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Nicole Scholz
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Bilge Güvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Nina Vardjan
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
21
|
Gonçalves-Ribeiro J, Savchak OK, Costa-Pinto S, Gomes JI, Rivas-Santisteban R, Lillo A, Sánchez Romero J, Sebastião AM, Navarrete M, Navarro G, Franco R, Vaz SH. Adenosine receptors are the on-and-off switch of astrocytic cannabinoid type 1 (CB1) receptor effect upon synaptic plasticity in the medial prefrontal cortex. Glia 2024; 72:1096-1116. [PMID: 38482984 DOI: 10.1002/glia.24518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 04/12/2024]
Abstract
The medial prefrontal cortex (mPFC) is involved in cognitive functions such as working memory. Astrocytic cannabinoid type 1 receptor (CB1R) induces cytosolic calcium (Ca2+) concentration changes with an impact on neuronal function. mPFC astrocytes also express adenosine A1 and A2A receptors (A1R, A2AR), being unknown the crosstalk between CB1R and adenosine receptors in these cells. We show here that a further level of regulation of astrocyte Ca2+ signaling occurs through CB1R-A2AR or CB1R-A1R heteromers that ultimately impact mPFC synaptic plasticity. CB1R-mediated Ca2+ transients increased and decreased when A1R and A2AR were activated, respectively, unveiling adenosine receptors as modulators of astrocytic CB1R. CB1R activation leads to an enhancement of long-term potentiation (LTP) in the mPFC, under the control of A1R but not of A2AR. Notably, in IP3R2KO mice, that do not show astrocytic Ca2+ level elevations, CB1R activation decreases LTP, which is not modified by A1R or A2AR. The present work suggests that CB1R has a homeostatic role on mPFC LTP, under the control of A1R, probably due to physical crosstalk between these receptors in astrocytes that ultimately alters CB1R Ca2+ signaling.
Collapse
Affiliation(s)
- Joana Gonçalves-Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Oksana K Savchak
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Costa-Pinto
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Joana I Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rafael Rivas-Santisteban
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Javier Sánchez Romero
- Instituto Cajal, CSIC, Madrid, Spain
- PhD Program in Neuroscience, Universidad Autónoma de Madrid-Instituto Cajal, Madrid, Spain
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | - Gemma Navarro
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Franco
- CiberNed, Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
22
|
Li Z, Jiang YY, Long C, Peng X, Tao J, Pu Y, Yue R. Bridging metabolic syndrome and cognitive dysfunction: role of astrocytes. Front Endocrinol (Lausanne) 2024; 15:1393253. [PMID: 38800473 PMCID: PMC11116704 DOI: 10.3389/fendo.2024.1393253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Metabolic syndrome (MetS) and cognitive dysfunction pose significant challenges to global health and the economy. Systemic inflammation, endocrine disruption, and autoregulatory impairment drive neurodegeneration and microcirculatory damage in MetS. Due to their unique anatomy and function, astrocytes sense and integrate multiple metabolic signals, including peripheral endocrine hormones and nutrients. Astrocytes and synapses engage in a complex dialogue of energetic and immunological interactions. Astrocytes act as a bridge between MetS and cognitive dysfunction, undergoing diverse activation in response to metabolic dysfunction. This article summarizes the alterations in astrocyte phenotypic characteristics across multiple pathological factors in MetS. It also discusses the clinical value of astrocytes as a critical pathologic diagnostic marker and potential therapeutic target for MetS-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Zihan Li
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ya-yi Jiang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Caiyi Long
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xi Peng
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiajing Tao
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueheng Pu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rensong Yue
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
23
|
Cahill MK, Collard M, Tse V, Reitman ME, Etchenique R, Kirst C, Poskanzer KE. Network-level encoding of local neurotransmitters in cortical astrocytes. Nature 2024; 629:146-153. [PMID: 38632406 PMCID: PMC11062919 DOI: 10.1038/s41586-024-07311-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024]
Abstract
Astrocytes, the most abundant non-neuronal cell type in the mammalian brain, are crucial circuit components that respond to and modulate neuronal activity through calcium (Ca2+) signalling1-7. Astrocyte Ca2+ activity is highly heterogeneous and occurs across multiple spatiotemporal scales-from fast, subcellular activity3,4 to slow, synchronized activity across connected astrocyte networks8-10-to influence many processes5,7,11. However, the inputs that drive astrocyte network dynamics remain unclear. Here we used ex vivo and in vivo two-photon astrocyte imaging while mimicking neuronal neurotransmitter inputs at multiple spatiotemporal scales. We find that brief, subcellular inputs of GABA and glutamate lead to widespread, long-lasting astrocyte Ca2+ responses beyond an individual stimulated cell. Further, we find that a key subset of Ca2+ activity-propagative activity-differentiates astrocyte network responses to these two main neurotransmitters, and may influence responses to future inputs. Together, our results demonstrate that local, transient neurotransmitter inputs are encoded by broad cortical astrocyte networks over a minutes-long time course, contributing to accumulating evidence that substantial astrocyte-neuron communication occurs across slow, network-level spatiotemporal scales12-14. These findings will enable future studies to investigate the link between specific astrocyte Ca2+ activity and specific functional outputs, which could build a consistent framework for astrocytic modulation of neuronal activity.
Collapse
Affiliation(s)
- Michelle K Cahill
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Max Collard
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Vincent Tse
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
| | - Michael E Reitman
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Roberto Etchenique
- Departamento de Química Inorgánica, Analítica y Química Física, INQUIMAE, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Christoph Kirst
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Kira E Poskanzer
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA.
| |
Collapse
|
24
|
Du C, Park K, Hua Y, Liu Y, Volkow ND, Pan Y. Astrocytes modulate cerebral blood flow and neuronal response to cocaine in prefrontal cortex. Mol Psychiatry 2024; 29:820-834. [PMID: 38238549 PMCID: PMC11784220 DOI: 10.1038/s41380-023-02373-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 01/30/2024]
Abstract
Cocaine affects both cerebral blood vessels and neuronal activity in brain. Cocaine can also disrupt astrocytes, which modulate neurovascular coupling-a process that regulates cerebral hemodynamics in response to neuronal activation. However, separating neuronal and astrocytic effects from cocaine's direct vasoactive effects has been challenging, partially due to limitations of neuroimaging techniques able to differentiate vascular from neuronal and glial effects at high temporal and spatial resolutions. Here, we used a newly-developed multi-channel fluorescence and optical coherence Doppler microscope (fl-ODM) that allows for simultaneous measurements of neuronal and astrocytic activities (reflected by the intracellular calcium changes in neurons Ca2+N and astrocytes Ca2+A, respectively) alongside their vascular interactions in vivo to address this challenge. Using green and red genetically-encoded Ca2+ indicators differentially expressed in astrocytes and neurons, fl-ODM enabled concomitant imaging of large-scale astrocytic and neuronal Ca2+ fluorescence and 3D cerebral blood flow velocity (CBFv) in vascular networks in the mouse cortex. We assessed cocaine's effects in the prefrontal cortex (PFC) and found that the CBFv changes triggered by cocaine were temporally correlated with astrocytic Ca2+A activity. Chemogenetic inhibition of astrocytes during the baseline state resulted in blood vessel dilation and CBFv increases but did not affect neuronal activity, suggesting modulation of spontaneous blood vessel's vascular tone by astrocytes. Chemogenetic inhibition of astrocytes during a cocaine challenge prevented its vasoconstricting effects alongside the CBFv decreases, but it also attenuated the neuronal Ca2+N increases triggered by cocaine. These results document a role of astrocytes both in regulating vascular tone and consequently blood flow, at baseline and for modulating the vasoconstricting and neuronal activation responses to cocaine in the PFC. Strategies to inhibit astrocytic activity could offer promise for ameliorating vascular and neuronal toxicity from cocaine misuse.
Collapse
Affiliation(s)
- Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Kichon Park
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Yueming Hua
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Yanzuo Liu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20857, USA
| | - Yingtian Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
25
|
Chen X, Tang SJ. Neural Circuitry Polarization in the Spinal Dorsal Horn (SDH): A Novel Form of Dysregulated Circuitry Plasticity during Pain Pathogenesis. Cells 2024; 13:398. [PMID: 38474361 PMCID: PMC10930392 DOI: 10.3390/cells13050398] [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: 01/20/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Pathological pain emerges from nociceptive system dysfunction, resulting in heightened pain circuit activity. Various forms of circuitry plasticity, such as central sensitization, synaptic plasticity, homeostatic plasticity, and excitation/inhibition balance, contribute to the malfunction of neural circuits during pain pathogenesis. Recently, a new form of plasticity in the spinal dorsal horn (SDH), named neural circuit polarization (NCP), was discovered in pain models induced by HIV-1 gp120 and chronic morphine administration. NCP manifests as an increase in excitatory postsynaptic currents (EPSCs) in excitatory neurons and a decrease in EPSCs in inhibitory neurons, presumably facilitating hyperactivation of pain circuits. The expression of NCP is associated with astrogliosis. Ablation of reactive astrocytes or suppression of astrogliosis blocks NCP and, concomitantly, the development of gp120- or morphine-induced pain. In this review, we aim to compare and integrate NCP with other forms of plasticity in pain circuits to improve the understanding of the pathogenic contribution of NCP and its cooperation with other forms of circuitry plasticity during the development of pathological pain.
Collapse
Affiliation(s)
| | - Shao-Jun Tang
- Stony Brook University Pain and Anesthesia Research Center (SPARC), Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA;
| |
Collapse
|
26
|
Rivera-Villaseñor A, Higinio-Rodríguez F, López-Hidalgo M. Astrocytes in Pain Perception: A Systems Neuroscience Approach. ADVANCES IN NEUROBIOLOGY 2024; 39:193-212. [PMID: 39190076 DOI: 10.1007/978-3-031-64839-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Astrocytes play an active role in the function of the brain integrating neuronal activity and regulating back neuronal dynamic. They have recently emerged as active contributors of brain's emergent properties such as perceptions. Here, we analyzed the role of astrocytes in pain perception from the lens of systems neuroscience, and we do this by analyzing how astrocytes encode nociceptive information within brain processing areas and how they are key regulators of the internal state that determines pain perception. Specifically, we discuss the dynamic interactions between astrocytes and neuromodulators, such as noradrenaline, highlighting their role in shaping the level of activation of the neuronal ensemble, thereby influencing the experience of pain. Also, we will discuss the possible implications of an "Astro-NeuroMatrix" in the integration of pain across sensory, affective, and cognitive dimensions of pain perception.
Collapse
Affiliation(s)
- Angélica Rivera-Villaseñor
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico
| | - Frida Higinio-Rodríguez
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico
| | - Mónica López-Hidalgo
- Escuela Nacional de Estudios Superiores, Universidad Nacional Autónoma de México, Queretaro, Qro., Mexico.
| |
Collapse
|
27
|
Cahill MK, Collard M, Tse V, Reitman ME, Etchenique R, Kirst C, Poskanzer KE. Network-level encoding of local neurotransmitters in cortical astrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.568932. [PMID: 38106119 PMCID: PMC10723263 DOI: 10.1101/2023.12.01.568932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Astrocytes-the most abundant non-neuronal cell type in the mammalian brain-are crucial circuit components that respond to and modulate neuronal activity via calcium (Ca 2+ ) signaling 1-8 . Astrocyte Ca 2+ activity is highly heterogeneous and occurs across multiple spatiotemporal scales: from fast, subcellular activity 3,4 to slow, synchronized activity that travels across connected astrocyte networks 9-11 . Furthermore, astrocyte network activity has been shown to influence a wide range of processes 5,8,12 . While astrocyte network activity has important implications for neuronal circuit function, the inputs that drive astrocyte network dynamics remain unclear. Here we used ex vivo and in vivo two-photon Ca 2+ imaging of astrocytes while mimicking neuronal neurotransmitter inputs at multiple spatiotemporal scales. We find that brief, subcellular inputs of GABA and glutamate lead to widespread, long-lasting astrocyte Ca 2+ responses beyond an individual stimulated cell. Further, we find that a key subset of Ca 2+ activity-propagative events-differentiates astrocyte network responses to these two major neurotransmitters, and gates responses to future inputs. Together, our results demonstrate that local, transient neurotransmitter inputs are encoded by broad cortical astrocyte networks over the course of minutes, contributing to accumulating evidence across multiple model organisms that significant astrocyte-neuron communication occurs across slow, network-level spatiotemporal scales 13-15 . We anticipate that this study will be a starting point for future studies investigating the link between specific astrocyte Ca 2+ activity and specific astrocyte functional outputs, which could build a consistent framework for astrocytic modulation of neuronal activity.
Collapse
|
28
|
Hjukse JB, Puebla MFDL, Vindedal GF, Sprengel R, Jensen V, Nagelhus EA, Tang W. Increased membrane Ca 2+ permeability drives astrocytic Ca 2+ dynamics during neuronal stimulation at excitatory synapses. Glia 2023; 71:2770-2781. [PMID: 37564028 DOI: 10.1002/glia.24450] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/13/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
Astrocytes are intricately involved in the activity of neural circuits; however, their basic physiology of interacting with nearby neurons is not well established. Using two-photon imaging of neurons and astrocytes during higher frequency stimulation of hippocampal CA3-CA1 Schaffer collateral (Scc) excitatory synapses, we could show that increasing levels of released glutamate accelerated local astrocytic Ca2+ elevation. However, blockage of glutamate transporters did not abolish this astrocytic Ca2+ response, suggesting that astrocytic Ca2+ elevation is indirectly associated with an uptake of extracellular glutamate. However, during the astrocytic glutamate uptake, the Na+ /Ca2+ exchanger (NCX) reverse mode was activated, and mediated extracellular Ca2+ entry, thereby triggering the internal release of Ca2+ . In addition, extracellular Ca2+ entry via membrane P2X receptors further facilitated astrocytic Ca2+ elevation via ATP binding. These findings suggest a novel mechanism of activity induced Ca2+ permeability increases of astrocytic membranes, which drives astrocytic responses during neuronal stimulation of CA3-CA1 Scc excitatory synapses.
Collapse
Affiliation(s)
- Jarand B Hjukse
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Mario F D L Puebla
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Neurology, Neuroclinic, St. Olavs Hospital, Trondheim, Norway
| | - Gry Fluge Vindedal
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Rolf Sprengel
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Vidar Jensen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Erlend A Nagelhus
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Research Group of Molecular Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Wannan Tang
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Neurology, Neuroclinic, St. Olavs Hospital, Trondheim, Norway
| |
Collapse
|
29
|
Vaiana AM, Chen Y, Gelfond J, Johnson-Pais TL, Leach RJ, Ramamurthy C, Thompson IM, Morilak DA. Effects of vortioxetine on hippocampal-related cognitive impairment induced in rats by androgen deprivation as a model of prostate cancer treatment. Transl Psychiatry 2023; 13:307. [PMID: 37788996 PMCID: PMC10547695 DOI: 10.1038/s41398-023-02600-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
Advances in prostate cancer treatment have significantly improved survival, but quality of life for survivors remains an under-studied area of research. Androgen deprivation therapy (ADT) is a foundational treatment for advanced prostate cancer and is used as an adjuvant for prolonged periods in many high-risk, localized tumors. More than half of patients treated with ADT experience debilitating cognitive impairments in domains such as spatial learning and working memory. In this study, we investigated the effects of androgen deprivation on hippocampal-mediated cognition in rats. Vortioxetine, a multimodal antidepressant, has been shown to improve cognition in depressed patients. Thus, we also tested the potential efficacy of vortioxetine in restoring impaired cognition after ADT. We further investigated mechanisms that might contribute to these effects, measuring changes in the circuitry and gene expression within the dorsal hippocampus. ADT via surgical castration induced impairments in visuospatial cognition on the novel object location test and attenuated afferent-evoked local field potentials recorded in the CA1 region of the dorsal hippocampus. Chronic dietary administration of vortioxetine effectively reversed these deficits. Castration significantly altered gene expression in the hippocampus, whereas vortioxetine had little effect. Pathway analysis revealed that androgen depletion altered pathways related to synaptic plasticity. These results suggest that the hippocampus may be vulnerable to ADT, contributing to cognitive impairment in prostate cancer patients. Further, vortioxetine may be a candidate to improve cognition in patients who experience cognitive decline after androgen deprivation therapy for prostate cancer and may do so by restoring molecular and circuit-level plasticity-related mechanisms compromised by ADT.
Collapse
Affiliation(s)
- Alexandra M Vaiana
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Population Health Sciences, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Jonathan Gelfond
- Department of Population Health Sciences, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Teresa L Johnson-Pais
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Urology, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Robin J Leach
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Cell Systems & Anatomy, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Chethan Ramamurthy
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Ian M Thompson
- Department of Urology, University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - David A Morilak
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
- Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
- Mays Cancer Center, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
- South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
| |
Collapse
|
30
|
González-Arias C, Sánchez-Ruiz A, Esparza J, Sánchez-Puelles C, Arancibia L, Ramírez-Franco J, Gobbo D, Kirchhoff F, Perea G. Dysfunctional serotonergic neuron-astrocyte signaling in depressive-like states. Mol Psychiatry 2023; 28:3856-3873. [PMID: 37773446 PMCID: PMC10730416 DOI: 10.1038/s41380-023-02269-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 10/01/2023]
Abstract
Astrocytes play crucial roles in brain homeostasis and are regulatory elements of neuronal and synaptic physiology. Astrocytic alterations have been found in Major Depressive Disorder (MDD) patients; however, the consequences of astrocyte Ca2+ signaling in MDD are poorly understood. Here, we found that corticosterone-treated juvenile mice (Cort-mice) showed altered astrocytic Ca2+ dynamics in mPFC both in resting conditions and during social interactions, in line with altered mice behavior. Additionally, Cort-mice displayed reduced serotonin (5-HT)-mediated Ca2+ signaling in mPFC astrocytes, and aberrant 5-HT-driven synaptic plasticity in layer 2/3 mPFC neurons. Downregulation of astrocyte Ca2+ signaling in naïve animals mimicked the synaptic deficits found in Cort-mice. Remarkably, boosting astrocyte Ca2+ signaling with Gq-DREADDS restored to the control levels mood and cognitive abilities in Cort-mice. This study highlights the important role of astrocyte Ca2+ signaling for homeostatic control of brain circuits and behavior, but also reveals its potential therapeutic value for depressive-like states.
Collapse
Affiliation(s)
- Candela González-Arias
- Cajal Institute, CSIC, 28002, Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University-Cajal Institute, Madrid, 28029, Spain
| | - Andrea Sánchez-Ruiz
- Cajal Institute, CSIC, 28002, Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University-Cajal Institute, Madrid, 28029, Spain
| | | | | | | | - Jorge Ramírez-Franco
- Institut de Neurosciences de la Timone, Aix-Marseille Université (AMU) & CNRS, UMR7289, 13005, Marseille, France
| | - Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421, Homburg, Germany
| | | |
Collapse
|
31
|
Noh K, Cho WH, Lee BH, Kim DW, Kim YS, Park K, Hwang M, Barcelon E, Cho YK, Lee CJ, Yoon BE, Choi SY, Park HY, Jun SB, Lee SJ. Cortical astrocytes modulate dominance behavior in male mice by regulating synaptic excitatory and inhibitory balance. Nat Neurosci 2023; 26:1541-1554. [PMID: 37563296 DOI: 10.1038/s41593-023-01406-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/17/2023] [Indexed: 08/12/2023]
Abstract
Social hierarchy is established as an outcome of individual social behaviors, such as dominance behavior during long-term interactions with others. Astrocytes are implicated in optimizing the balance between excitatory and inhibitory (E/I) neuronal activity, which may influence social behavior. However, the contribution of astrocytes in the prefrontal cortex to dominance behavior is unclear. Here we show that dorsomedial prefrontal cortical (dmPFC) astrocytes modulate E/I balance and dominance behavior in adult male mice using in vivo fiber photometry and two-photon microscopy. Optogenetic and chemogenetic activation or inhibition of dmPFC astrocytes show that astrocytes bidirectionally control male mouse dominance behavior, affecting social rank. Dominant and subordinate male mice present distinct prefrontal synaptic E/I balance, regulated by astrocyte activity. Mechanistically, we show that dmPFC astrocytes control cortical E/I balance by simultaneously enhancing presynaptic-excitatory and reducing postsynaptic-inhibitory transmission via astrocyte-derived glutamate and ATP release, respectively. Our findings show how dmPFC astrocyte-neuron communication can be involved in the establishment of social hierarchy in adult male mice.
Collapse
Affiliation(s)
- Kyungchul Noh
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Woo-Hyun Cho
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Byung Hun Lee
- Department of Physics and Astronomy, Seoul National University, Seoul, Republic of Korea
| | - Dong Wook Kim
- Department of Physics and Astronomy, Seoul National University, Seoul, Republic of Korea
| | - Yoo Sung Kim
- Department of Molecular Biology, Dankook University, Cheonan, Republic of Korea
| | - Keebum Park
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Minkyu Hwang
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Ellane Barcelon
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Yoon Kyung Cho
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Bo-Eun Yoon
- Department of Molecular Biology, Dankook University, Cheonan, Republic of Korea
| | - Se-Young Choi
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Hye Yoon Park
- Department of Physics and Astronomy, Seoul National University, Seoul, Republic of Korea
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Sang Beom Jun
- Department of Electronic and Electrical Engineering, Ewha Womans University, Seoul, Republic of Korea
- Graduate Program in Smart Factory, Ewha Womans University, Seoul, Republic of Korea
- Department of Brain & Cognitive Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Sung Joong Lee
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Ferré S, Köfalvi A, Ciruela F, Justinova Z, Pistis M. Targeting corticostriatal transmission for the treatment of cannabinoid use disorder. Trends Pharmacol Sci 2023; 44:495-506. [PMID: 37331914 PMCID: PMC10524660 DOI: 10.1016/j.tips.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/20/2023]
Abstract
It is generally assumed that the rewarding effects of cannabinoids are mediated by cannabinoid CB1 receptors (CB1Rs) the activation of which disinhibits dopaminergic neurons in the ventral tegmental area (VTA). However, this mechanism cannot fully explain novel results indicating that dopaminergic neurons also mediate the aversive effects of cannabinoids in rodents, and previous results showing that preferentially presynaptic adenosine A2A receptor (A2AR) antagonists counteract self-administration of Δ-9-tetrahydrocannabinol (THC) in nonhuman primates (NHPs). Based on recent experiments in rodents and imaging studies in humans, we propose that the activation of frontal corticostriatal glutamatergic transmission constitutes an additional and necessary mechanism. Here, we review evidence supporting the involvement of cortical astrocytic CB1Rs in the activation of corticostriatal neurons and that A2AR receptor heteromers localized in striatal glutamatergic terminals mediate the counteracting effects of the presynaptic A2AR antagonists, constituting potential targets for the treatment of cannabinoid use disorder (CUD).
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| | - Attila Köfalvi
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain; Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Zuzana Justinova
- Division of Pharmacology, Physiology, and Biological Chemistry (PPBC), National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Marco Pistis
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy
| |
Collapse
|
33
|
Jin Y, Song D, Yan Y, Quan Z, Qing H. The Role of Oxytocin in Early-Life-Stress-Related Neuropsychiatric Disorders. Int J Mol Sci 2023; 24:10430. [PMID: 37445607 DOI: 10.3390/ijms241310430] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Early-life stress during critical periods of brain development can have long-term effects on physical and mental health. Oxytocin is a critical social regulator and anti-inflammatory hormone that modulates stress-related functions and social behaviors and alleviates diseases. Oxytocin-related neural systems show high plasticity in early postpartum and adolescent periods. Early-life stress can influence the oxytocin system long term by altering the expression and signaling of oxytocin receptors. Deficits in social behavior, emotional control, and stress responses may result, thus increasing the risk of anxiety, depression, and other stress-related neuropsychiatric diseases. Oxytocin is regarded as an important target for the treatment of stress-related neuropsychiatric disorders. Here, we describe the history of oxytocin and its role in neural circuits and related behaviors. We then review abnormalities in the oxytocin system in early-life stress and the functions of oxytocin in treating stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yue Jin
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Da Song
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
34
|
Purushotham SS, Buskila Y. Astrocytic modulation of neuronal signalling. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1205544. [PMID: 37332623 PMCID: PMC10269688 DOI: 10.3389/fnetp.2023.1205544] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023]
Abstract
Neuronal signalling is a key element in neuronal communication and is essential for the proper functioning of the CNS. Astrocytes, the most prominent glia in the brain play a key role in modulating neuronal signalling at the molecular, synaptic, cellular, and network levels. Over the past few decades, our knowledge about astrocytes and their functioning has evolved from considering them as merely a brain glue that provides structural support to neurons, to key communication elements. Astrocytes can regulate the activity of neurons by controlling the concentrations of ions and neurotransmitters in the extracellular milieu, as well as releasing chemicals and gliotransmitters that modulate neuronal activity. The aim of this review is to summarise the main processes through which astrocytes are modulating brain function. We will systematically distinguish between direct and indirect pathways in which astrocytes affect neuronal signalling at all levels. Lastly, we will summarize pathological conditions that arise once these signalling pathways are impaired focusing on neurodegeneration.
Collapse
Affiliation(s)
| | - Yossi Buskila
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- The MARCS Institute, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
35
|
Andrade-Talavera Y, Fisahn A, Rodríguez-Moreno A. Timing to be precise? An overview of spike timing-dependent plasticity, brain rhythmicity, and glial cells interplay within neuronal circuits. Mol Psychiatry 2023; 28:2177-2188. [PMID: 36991134 PMCID: PMC10611582 DOI: 10.1038/s41380-023-02027-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/31/2023]
Abstract
In the mammalian brain information processing and storage rely on the complex coding and decoding events performed by neuronal networks. These actions are based on the computational ability of neurons and their functional engagement in neuronal assemblies where precise timing of action potential firing is crucial. Neuronal circuits manage a myriad of spatially and temporally overlapping inputs to compute specific outputs that are proposed to underly memory traces formation, sensory perception, and cognitive behaviors. Spike-timing-dependent plasticity (STDP) and electrical brain rhythms are suggested to underlie such functions while the physiological evidence of assembly structures and mechanisms driving both processes continues to be scarce. Here, we review foundational and current evidence on timing precision and cooperative neuronal electrical activity driving STDP and brain rhythms, their interactions, and the emerging role of glial cells in such processes. We also provide an overview of their cognitive correlates and discuss current limitations and controversies, future perspectives on experimental approaches, and their application in humans.
Collapse
Affiliation(s)
- Yuniesky Andrade-Talavera
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, ES-41013, Seville, Spain.
| | - André Fisahn
- Department of Biosciences and Nutrition and Department of Women's and Children's Health, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Antonio Rodríguez-Moreno
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, Universidad Pablo de Olavide, ES-41013, Seville, Spain.
| |
Collapse
|
36
|
Hashimoto A, Kawamura N, Tarusawa E, Takeda I, Aoyama Y, Ohno N, Inoue M, Kagamiuchi M, Kato D, Matsumoto M, Hasegawa Y, Nabekura J, Schaefer A, Moorhouse AJ, Yagi T, Wake H. Microglia enable cross-modal plasticity by removing inhibitory synapses. Cell Rep 2023; 42:112383. [PMID: 37086724 DOI: 10.1016/j.celrep.2023.112383] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/13/2023] [Accepted: 03/28/2023] [Indexed: 04/24/2023] Open
Abstract
Cross-modal plasticity is the repurposing of brain regions associated with deprived sensory inputs to improve the capacity of other sensory modalities. The functional mechanisms of cross-modal plasticity can indicate how the brain recovers from various forms of injury and how different sensory modalities are integrated. Here, we demonstrate that rewiring of the microglia-mediated local circuit synapse is crucial for cross-modal plasticity induced by visual deprivation (monocular deprivation [MD]). MD relieves the usual inhibition of functional connectivity between the somatosensory cortex and secondary lateral visual cortex (V2L). This results in enhanced excitatory responses in V2L neurons during whisker stimulation and a greater capacity for vibrissae sensory discrimination. The enhanced cross-modal response is mediated by selective removal of inhibitory synapse terminals on pyramidal neurons by the microglia in the V2L via matrix metalloproteinase 9 signaling. Our results provide insights into how cortical circuits integrate different inputs to functionally compensate for neuronal damage.
Collapse
Affiliation(s)
- Akari Hashimoto
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Nanami Kawamura
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Etsuko Tarusawa
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Ikuko Takeda
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yuki Aoyama
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, Shimotsuke 329-0498, Japan; Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Mio Inoue
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mai Kagamiuchi
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mami Matsumoto
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Yoshihiro Hasegawa
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; Department of Physiological Sciences, Graduate University for Advanced Studies, SOKENDAI, Hayama 240-0193, Japan
| | - Anne Schaefer
- Center for Glial Biology, Department of Neuroscience and Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA; Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Andrew J Moorhouse
- School of Biomedical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Takeshi Yagi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; Center for Optical Scattering Image Science, Kobe University, Kobe 657-8501, Japan; Department of Physiological Sciences, Graduate University for Advanced Studies, SOKENDAI, Hayama 240-0193, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan.
| |
Collapse
|
37
|
Nanclares C, Poynter J, Martell-Martinez HA, Vermilyea S, Araque A, Kofuji P, Lee MK, Covelo A. Dysregulation of astrocytic Ca 2+ signaling and gliotransmitter release in mouse models of α-synucleinopathies. Acta Neuropathol 2023; 145:597-610. [PMID: 36764943 PMCID: PMC10119048 DOI: 10.1007/s00401-023-02547-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/17/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023]
Abstract
α-Synuclein is a major component of Lewy bodies (LB) and Lewy neurites (LN) appearing in the postmortem brain of Parkinson's disease (PD) and other α-synucleinopathies. While most studies of α-synucleinopathies have focused on neuronal and synaptic alterations as well as dysfunctions of the astrocytic homeostatic roles, whether the bidirectional astrocyte-neuronal communication is affected in these diseases remains unknown. We have investigated whether the astrocyte Ca2+ excitability and the glutamatergic gliotransmission underlying astrocyte-neuronal signaling are altered in several transgenic mouse models related to α-synucleinopathies, i.e., mice expressing high and low levels of the human A53T mutant α-synuclein (G2-3 and H5 mice, respectively) globally or selectively in neurons (iSyn mice), mice expressing human wildtype α-synuclein (I2-2 mice), and mice expressing A30P mutant α-synuclein (O2 mice). Combining astrocytic Ca2+ imaging and neuronal electrophysiological recordings in hippocampal slices of these mice, we have found that compared to non-transgenic mice, astrocytes in G2-3 mice at different ages (1-6 months) displayed a Ca2+ hyperexcitability that was independent of neurotransmitter receptor activation, suggesting that the expression of α-synuclein mutant A53T altered the intrinsic properties of astrocytes. Similar dysregulation of the astrocyte Ca2+ signal was present in H5 mice, but not in I2-2 and O2 mice, indicating α-synuclein mutant-specific effects. Moreover, astrocyte Ca2+ hyperexcitability was absent in mice expressing the α-synuclein mutant A53T selectively in neurons, indicating that the effects on astrocytes were cell-autonomous. Consistent with these effects, glutamatergic gliotransmission was enhanced in G2-3 and H5 mice, but was unaffected in I2-2, O2 and iSyn mice. These results indicate a cell-autonomous effect of pathogenic A53T expression in astrocytes that may contribute to the altered neuronal and synaptic function observed in α-synucleinopathies.
Collapse
Affiliation(s)
- Carmen Nanclares
- Department of Neuroscience, University of Minnesota, 4-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Jonah Poynter
- Department of Neuroscience, University of Minnesota, 4-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Hector A Martell-Martinez
- Department of Neuroscience, University of Minnesota, 4-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Scott Vermilyea
- Department of Neuroscience, University of Minnesota, 4-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, 4-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, 4-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
| | - Michael K Lee
- Department of Neuroscience, University of Minnesota, 4-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN, 55455, USA.
| | - Ana Covelo
- Department of Neuroscience, University of Minnesota, 4-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, 33077, Bordeaux, France.
- University of Bordeaux, 33077, Bordeaux, France.
| |
Collapse
|
38
|
Hwang Y, Park JH, Kim HC, Shin EJ. GABA B receptor activation alters astrocyte phenotype changes induced by trimethyltin via ERK signaling in the dentate gyrus of mice. Life Sci 2023; 319:121529. [PMID: 36841471 DOI: 10.1016/j.lfs.2023.121529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/11/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023]
Abstract
AIMS We examined the effect of γ-aminobutyric acid (GABA)B receptor activation on astrocyte phenotype changes induced by trimethyltin (TMT) in the dentate gyrus of mice. MAIN METHODS Male C57BL/6N mice received TMT (2.6 mg/kg, i.p.), and the expression of GABAB receptors was evaluated in the hippocampus. The GABAB receptor agonist baclofen (2.5, 5, or 10 mg/kg, i.p. × 5 at 12-h intervals) was administered 3-5 days after TMT treatment, and the expression of Iba-1, GFAP, and astrocyte phenotype markers was evaluated 6 days after TMT. SL327 (30 mg/kg, i.p.), an extracellular signal-related kinase (ERK) inhibitor, was administered 1 h after each baclofen treatment. KEY FINDINGS TMT insult significantly induced the astroglial expression of GABAB receptors in the dentate molecular layer. Baclofen significantly promoted the expression of S100A10, EMP1, and CD109, but not that of C3, GGTA1, and MX1 induced by TMT. In addition, baclofen significantly increased the TMT-induced expression of p-ERK in the dentate molecular layer. Interestingly, p-ERK was more colocalized with S100A10 than with C3 after TMT insult, and a significant positive correlation was found between the expression of p-ERK and S100A10. Consistently, SL327 reversed the effect of baclofen on astrocyte phenotype changes. Baclofen also enhanced the TMT-induced astroglial expression of glial cell-derived neurotrophic factor (GDNF), an anti-inflammatory astrocytes-to-microglia mediator, and consequently attenuated Iba-1 expression and delayed apoptotic neuronal death. SIGNIFICANCE Our results suggest that GABAB receptor activation increases S100A10-positive anti-inflammatory astrocytes and astroglial GDNF expression via ERK signaling after TMT excitotoxicity in the dentate molecular layer of mice.
Collapse
Affiliation(s)
- Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jung Hoon Park
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
39
|
Dzyubenko E, Hermann DM. Role of glia and extracellular matrix in controlling neuroplasticity in the central nervous system. Semin Immunopathol 2023:10.1007/s00281-023-00989-1. [PMID: 37052711 DOI: 10.1007/s00281-023-00989-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/24/2023] [Indexed: 04/14/2023]
Abstract
Neuronal plasticity is critical for the maintenance and modulation of brain activity. Emerging evidence indicates that glial cells actively shape neuroplasticity, allowing for highly flexible regulation of synaptic transmission, neuronal excitability, and network synchronization. Astrocytes regulate synaptogenesis, stabilize synaptic connectivity, and preserve the balance between excitation and inhibition in neuronal networks. Microglia, the brain-resident immune cells, continuously monitor and sculpt synapses, allowing for the remodeling of brain circuits. Glia-mediated neuroplasticity is driven by neuronal activity, controlled by a plethora of feedback signaling mechanisms and crucially involves extracellular matrix remodeling in the central nervous system. This review summarizes the key findings considering neurotransmission regulation and metabolic support by astrocyte-neuronal networks, and synaptic remodeling mediated by microglia. Novel data indicate that astrocytes and microglia are pivotal for controlling brain function, indicating the necessity to rethink neurocentric neuroplasticity views.
Collapse
Affiliation(s)
- Egor Dzyubenko
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Dirk M Hermann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
40
|
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
|
41
|
Manninen T, Aćimović J, Linne ML. Analysis of Network Models with Neuron-Astrocyte Interactions. Neuroinformatics 2023; 21:375-406. [PMID: 36959372 PMCID: PMC10085960 DOI: 10.1007/s12021-023-09622-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2023] [Indexed: 03/25/2023]
Abstract
Neural networks, composed of many neurons and governed by complex interactions between them, are a widely accepted formalism for modeling and exploring global dynamics and emergent properties in brain systems. In the past decades, experimental evidence of computationally relevant neuron-astrocyte interactions, as well as the astrocytic modulation of global neural dynamics, have accumulated. These findings motivated advances in computational glioscience and inspired several models integrating mechanisms of neuron-astrocyte interactions into the standard neural network formalism. These models were developed to study, for example, synchronization, information transfer, synaptic plasticity, and hyperexcitability, as well as classification tasks and hardware implementations. We here focus on network models of at least two neurons interacting bidirectionally with at least two astrocytes that include explicitly modeled astrocytic calcium dynamics. In this study, we analyze the evolution of these models and the biophysical, biochemical, cellular, and network mechanisms used to construct them. Based on our analysis, we propose how to systematically describe and categorize interaction schemes between cells in neuron-astrocyte networks. We additionally study the models in view of the existing experimental data and present future perspectives. Our analysis is an important first step towards understanding astrocytic contribution to brain functions. However, more advances are needed to collect comprehensive data about astrocyte morphology and physiology in vivo and to better integrate them in data-driven computational models. Broadening the discussion about theoretical approaches and expanding the computational tools is necessary to better understand astrocytes' roles in brain functions.
Collapse
Affiliation(s)
- Tiina Manninen
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, FI-33720, Tampere, Finland.
| | - Jugoslava Aćimović
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, FI-33720, Tampere, Finland
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere University, Korkeakoulunkatu 3, FI-33720, Tampere, Finland.
| |
Collapse
|
42
|
Guidolin D, Tortorella C, Marcoli M, Cervetto C, Maura G, Agnati LF. Receptor-receptor interactions and microvesicle exchange as mechanisms modulating signaling between neurons and astrocytes. Neuropharmacology 2023; 231:109509. [PMID: 36935005 DOI: 10.1016/j.neuropharm.2023.109509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/21/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
It is well known that astrocytes play a significant metabolic role in the nervous tissue, maintaining the homeostasis of the extracellular space and of the blood-brain barrier, and providing trophic support to neurons. In addition, however, evidence exists indicating astrocytes as important elements for brain activity through signaling exchange with neurons. Astrocytes, indeed, can sense synaptic activity and their molecular machinery responds to neurotransmitters released by neurons with cytoplasmic Ca2+ elevations that, in turn, stimulate the release of neuroactive substances (gliotransmitters) influencing nearby neurons. In both cell types the recognition and transduction of this complex pattern of signals is mediated by specific receptors that are also involved in mechanisms tuning the intercellular cross-talk between astrocytes and neurons. Two of these mechanisms are the focus of the present discussion. The first concerns direct receptor-receptor interactions leading to the formation at the cell membrane of multimeric receptor complexes. The cooperativity that emerges in the actions of orthosteric and allosteric ligands of the monomers forming the assembly provides the cell decoding apparatus with sophisticated and flexible dynamics in terms of recognition and signal transduction pathways. A further mechanism of plasticity involving receptors is based on the transfer of elements of the cellular signaling apparatus via extracellular microvesicles acting as protective containers, which can lead to transient changes in the transmitting/decoding capabilities of the target cell.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121, Padova, Italy.
| | - Cinzia Tortorella
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121, Padova, Italy
| | - Manuela Marcoli
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126, Genova, Italy
| | - Chiara Cervetto
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126, Genova, Italy
| | - Guido Maura
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126, Genova, Italy
| | - Luigi F Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| |
Collapse
|
43
|
Goenaga J, Araque A, Kofuji P, Herrera Moro Chao D. Calcium signaling in astrocytes and gliotransmitter release. Front Synaptic Neurosci 2023; 15:1138577. [PMID: 36937570 PMCID: PMC10017551 DOI: 10.3389/fnsyn.2023.1138577] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Glia are as numerous in the brain as neurons and widely known to serve supportive roles such as structural scaffolding, extracellular ionic and neurotransmitter homeostasis, and metabolic support. However, over the past two decades, several lines of evidence indicate that astrocytes, which are a type of glia, play active roles in neural information processing. Astrocytes, although not electrically active, can exhibit a form of excitability by dynamic changes in intracellular calcium levels. They sense synaptic activity and release neuroactive substances, named gliotransmitters, that modulate neuronal activity and synaptic transmission in several brain areas, thus impacting animal behavior. This "dialogue" between astrocytes and neurons is embodied in the concept of the tripartite synapse that includes astrocytes as integral elements of synaptic function. Here, we review the recent work and discuss how astrocytes via calcium-mediated excitability modulate synaptic information processing at various spatial and time scales.
Collapse
Affiliation(s)
| | | | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
44
|
Cuellar-Santoyo AO, Ruiz-Rodríguez VM, Mares-Barbosa TB, Patrón-Soberano A, Howe AG, Portales-Pérez DP, Miquelajáuregui Graf A, Estrada-Sánchez AM. Revealing the contribution of astrocytes to glutamatergic neuronal transmission. Front Cell Neurosci 2023; 16:1037641. [PMID: 36744061 PMCID: PMC9893894 DOI: 10.3389/fncel.2022.1037641] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Research on glutamatergic neurotransmission has focused mainly on the function of presynaptic and postsynaptic neurons, leaving astrocytes with a secondary role only to ensure successful neurotransmission. However, recent evidence indicates that astrocytes contribute actively and even regulate neuronal transmission at different levels. This review establishes a framework by comparing glutamatergic components between neurons and astrocytes to examine how astrocytes modulate or otherwise influence neuronal transmission. We have included the most recent findings about the role of astrocytes in neurotransmission, allowing us to understand the complex network of neuron-astrocyte interactions. However, despite the knowledge of synaptic modulation by astrocytes, their contribution to specific physiological and pathological conditions remains to be elucidated. A full understanding of the astrocyte's role in neuronal processing could open fruitful new frontiers in the development of therapeutic applications.
Collapse
Affiliation(s)
- Ares Orlando Cuellar-Santoyo
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Victor Manuel Ruiz-Rodríguez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Teresa Belem Mares-Barbosa
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | - Araceli Patrón-Soberano
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Andrew G. Howe
- Intelligent Systems Laboratory, HRL Laboratories, LLC, Malibu, CA, United States
| | - Diana Patricia Portales-Pérez
- Translational and Molecular Medicine Laboratory, Research Center for Health Sciences and Biomedicine, Autonomous University of San Luis Potosí, San Luis Potosí, Mexico
| | | | - Ana María Estrada-Sánchez
- División de Biología Molecular, Laboratorio de Neurobiología, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| |
Collapse
|
45
|
Maltsev A, Roshchin M, Bezprozvanny I, Smirnov I, Vlasova O, Balaban P, Borodinova A. Bidirectional regulation by "star forces": Ionotropic astrocyte's optical stimulation suppresses synaptic plasticity, metabotropic one strikes back. Hippocampus 2023; 33:18-36. [PMID: 36484471 DOI: 10.1002/hipo.23486] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022]
Abstract
The role of astrocytes in modulating synaptic plasticity is an important question that until recently was not addressed due to limitations of previously existing technology. In the present study, we took an advantage of optogenetics to specifically activate astrocytes in hippocampal slices in order to study effects on synaptic function. Using the AAV-based delivery strategy, we expressed the ionotropic channelrhodopsin-2 (ChR2) or the metabotropic Gq-coupled Opto-a1AR opsins specifically in hippocampal astrocytes to compare different modalities of astrocyte activation. In electrophysiological experiments, we observed a depression of basal field excitatory postsynaptic potentials (fEPSPs) in the CA1 hippocampal layer following light stimulation of astrocytic ChR2. The ChR2-mediated depression increased under simultaneous light and electrical theta-burst stimulation (TBS). Application of the type 2 purinergic receptor antagonist suramin prevented depression of basal synaptic transmission, and switched the ChR2-dependent depression into potentiation. The GABAB receptor antagonist, phaclofen, did not prevent the depression of basal fEPSPs, but switched the ChR2-dependent depression into potentiation comparable to the values for TBS in control slices. In contrast, light stimulation of Opto-a1AR expressed in astrocytes led to an increase in basal fEPSPs, as well as a potentiation of synaptic responses to TBS significantly. A specific blocker of the Gq protein downstream target, the phospholipase C, U73122, completely prevented the effects of Opto-a1AR stimulation on basal fEPSPs or Opto + TBS responses. To understand molecular basis for the observed effects, we performed an analysis of gene expression in these slices using quantitative PCR approach. We observed a significant upregulation of "immediate-early" gene expression in hippocampal slices after light activation of Opto-a1AR-expressing astrocytes alone (cRel, Arc, Fos, JunB, and Egr1) or paired with TBS (cRel, Fos, and Egr1). Activation of ChR2-expressing hippocampal astrocytes was insufficient to affect expression of these genes in our experimental conditions. Thus, we concluded that optostimulation of astrocytes with ChR2 and Opto-a1AR optogenetic tools enables bidirectional modulation of synaptic plasticity and gene expression in hippocampus.
Collapse
Affiliation(s)
- Alexander Maltsev
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Matvey Roshchin
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia.,Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Ivan Smirnov
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Olga Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Pavel Balaban
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia Borodinova
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
46
|
Kruyer A, Kalivas PW, Scofield MD. Astrocyte regulation of synaptic signaling in psychiatric disorders. Neuropsychopharmacology 2023; 48:21-36. [PMID: 35577914 PMCID: PMC9700696 DOI: 10.1038/s41386-022-01338-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 02/07/2023]
Abstract
Over the last 15 years, the field of neuroscience has evolved toward recognizing the critical role of astroglia in shaping neuronal synaptic activity and along with the pre- and postsynapse is now considered an equal partner in tripartite synaptic transmission and plasticity. The relative youth of this recognition and a corresponding deficit in reagents and technologies for quantifying and manipulating astroglia relative to neurons continues to hamper advances in understanding tripartite synaptic physiology. Nonetheless, substantial advances have been made and are reviewed herein. We review the role of astroglia in synaptic function and regulation of behavior with an eye on how tripartite synapses figure into brain pathologies underlying behavioral impairments in psychiatric disorders, both from the perspective of measures in postmortem human brains and more subtle influences on tripartite synaptic regulation of behavior in animal models of psychiatric symptoms. Our goal is to provide the reader a well-referenced state-of-the-art understanding of current knowledge and predict what we may discover with deeper investigation of tripartite synapses using reagents and technologies not yet available.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
47
|
Jiménez-Dinamarca I, Reyes-Lizana R, Lemunao-Inostroza Y, Cárdenas K, Castro-Lazo R, Peña F, Lucero CM, Prieto-Villalobos J, Retamal MA, Orellana JA, Stehberg J. GABAergic Regulation of Astroglial Gliotransmission through Cx43 Hemichannels. Int J Mol Sci 2022; 23:13625. [PMID: 36362410 PMCID: PMC9656947 DOI: 10.3390/ijms232113625] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/30/2022] [Accepted: 10/09/2022] [Indexed: 02/12/2024] Open
Abstract
Gamma-Aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the brain. It is produced by interneurons and recycled by astrocytes. In neurons, GABA activates the influx of Cl- via the GABAA receptor or efflux or K+ via the GABAB receptor, inducing hyperpolarization and synaptic inhibition. In astrocytes, the activation of both GABAA and GABAB receptors induces an increase in intracellular Ca2+ and the release of glutamate and ATP. Connexin 43 (Cx43) hemichannels are among the main Ca2+-dependent cellular mechanisms for the astroglial release of glutamate and ATP. However, no study has evaluated the effect of GABA on astroglial Cx43 hemichannel activity and Cx43 hemichannel-mediated gliotransmission. Here we assessed the effects of GABA on Cx43 hemichannel activity in DI NCT1 rat astrocytes and hippocampal brain slices. We found that GABA induces a Ca2+-dependent increase in Cx43 hemichannel activity in astrocytes mediated by the GABAA receptor, as it was blunted by the GABAA receptor antagonist bicuculline but unaffected by GABAB receptor antagonist CGP55845. Moreover, GABA induced the Cx43 hemichannel-dependent release of glutamate and ATP, which was also prevented by bicuculline, but unaffected by CGP. Gliotransmission in response to GABA was also unaffected by pannexin 1 channel blockade. These results are discussed in terms of the possible role of astroglial Cx43 hemichannel-mediated glutamate and ATP release in regulating the excitatory/inhibitory balance in the brain and their possible contribution to psychiatric disorders.
Collapse
Affiliation(s)
- Ivanka Jiménez-Dinamarca
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad Andres Bello, Santiago 8370186, Chile
| | - Rachel Reyes-Lizana
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad Andres Bello, Santiago 8370186, Chile
| | - Yordan Lemunao-Inostroza
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad Andres Bello, Santiago 8370186, Chile
| | - Kevin Cárdenas
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad Andres Bello, Santiago 8370186, Chile
| | - Raimundo Castro-Lazo
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad Andres Bello, Santiago 8370186, Chile
| | - Francisca Peña
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad Andres Bello, Santiago 8370186, Chile
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Universidad del Desarrollo–Clínica Alemana, Santiago 7780272, Chile
| | - Claudia M. Lucero
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Juan Prieto-Villalobos
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Mauricio Antonio Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Universidad del Desarrollo–Clínica Alemana, Santiago 7780272, Chile
| | - Juan Andrés Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Jimmy Stehberg
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad Andres Bello, Santiago 8370186, Chile
| |
Collapse
|
48
|
Multiple forms of working memory emerge from synapse-astrocyte interactions in a neuron-glia network model. Proc Natl Acad Sci U S A 2022; 119:e2207912119. [PMID: 36256810 PMCID: PMC9618090 DOI: 10.1073/pnas.2207912119] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Persistent activity in populations of neurons, time-varying activity across a neural population, or activity-silent mechanisms carried out by hidden internal states of the neural population have been proposed as different mechanisms of working memory (WM). Whether these mechanisms could be mutually exclusive or occur in the same neuronal circuit remains, however, elusive, and so do their biophysical underpinnings. While WM is traditionally regarded to depend purely on neuronal mechanisms, cortical networks also include astrocytes that can modulate neural activity. We propose and investigate a network model that includes both neurons and glia and show that glia-synapse interactions can lead to multiple stable states of synaptic transmission. Depending on parameters, these interactions can lead in turn to distinct patterns of network activity that can serve as substrates for WM.
Collapse
|
49
|
Cediel ML, Stawarski M, Blanc X, Nosková L, Magner M, Platzer K, Gburek-Augustat J, Baldridge D, Constantino JN, Ranza E, Bettler B, Antonarakis SE. GABBR1 monoallelic de novo variants linked to neurodevelopmental delay and epilepsy. Am J Hum Genet 2022; 109:1885-1893. [PMID: 36103875 PMCID: PMC9606381 DOI: 10.1016/j.ajhg.2022.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/15/2022] [Indexed: 01/25/2023] Open
Abstract
GABAB receptors are obligatory heterodimers responsible for prolonged neuronal inhibition in the central nervous system. The two receptor subunits are encoded by GABBR1 and GABBR2. Variants in GABBR2 have been associated with a Rett-like phenotype (MIM: 617903), epileptic encephalopathy (MIM: 617904), and milder forms of developmental delay with absence epilepsy. To date, however, no phenotypes associated with pathogenic variants of GABBR1 have been established. Through GeneMatcher, we have ascertained four individuals who each have a monoallelic GABBR1 de novo non-synonymous variant; these individuals exhibit motor and/or language delay, ranging from mild to severe, and in one case, epilepsy. Further phenotypic features include varying degrees of intellectual disability, learning difficulties, autism, ADHD, ODD, sleep disorders, and muscular hypotonia. We functionally characterized the four de novo GABBR1 variants, p.Glu368Asp, p.Ala397Val, p.Ala535Thr, and p.Gly673Asp, in transfected HEK293 cells. GABA fails to efficiently activate the variant receptors, most likely leading to an increase in the excitation/inhibition balance in the central nervous system. Variant p.Gly673Asp in transmembrane domain 3 (TMD3) renders the receptor completely inactive, consistent with failure of the receptor to reach the cell surface. p.Glu368Asp is located near the orthosteric binding site and reduces GABA potency and efficacy at the receptor. GABA exhibits normal potency but decreased efficacy at the p.Ala397Val and p.Ala535Thr variants. Functional characterization of GABBR1-related variants provides a rationale for understanding the severity of disease phenotypes and points to possible therapeutic strategies.
Collapse
Affiliation(s)
- Maria Lucia Cediel
- Medigenome, Swiss Institute of Genomic Medicine, 1207 Geneva, Switzerland
| | - Michal Stawarski
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Xavier Blanc
- Medigenome, Swiss Institute of Genomic Medicine, 1207 Geneva, Switzerland
| | - Lenka Nosková
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Martin Magner
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic; Department of Pediatrics, First Faculty of Medicine, Charles University and University Thomayer Hospital in Prague, Prague, Czech Republic
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Janina Gburek-Augustat
- Division of Neuropaediatrics, Hospital for Children and Adolescents, University Hospital Leipzig, Leipzig, Germany
| | | | | | - Emmanuelle Ranza
- Medigenome, Swiss Institute of Genomic Medicine, 1207 Geneva, Switzerland
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | | |
Collapse
|
50
|
Kruyer A. Astrocyte Heterogeneity in Regulation of Synaptic Activity. Cells 2022; 11:cells11193135. [PMID: 36231097 PMCID: PMC9562199 DOI: 10.3390/cells11193135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/02/2022] [Accepted: 10/02/2022] [Indexed: 02/07/2023] Open
Abstract
Our awareness of the number of synapse regulatory functions performed by astroglia is rapidly expanding, raising interesting questions regarding astrocyte heterogeneity and specialization across brain regions. Whether all astrocytes are poised to signal in a multitude of ways, or are instead tuned to surrounding synapses and how astroglial signaling is altered in psychiatric and cognitive disorders are fundamental questions for the field. In recent years, molecular and morphological characterization of astroglial types has broadened our ability to design studies to better analyze and manipulate specific functions of astroglia. Recent data emerging from these studies will be discussed in depth in this review. I also highlight remaining questions emerging from new techniques recently applied toward understanding the roles of astrocytes in synapse regulation in the adult brain.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|