1
|
Junaid M, Lee EJ, Lim SB. Single-cell and spatial omics: exploring hypothalamic heterogeneity. Neural Regen Res 2025; 20:1525-1540. [PMID: 38993130 PMCID: PMC11688568 DOI: 10.4103/nrr.nrr-d-24-00231] [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: 02/26/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
Elucidating the complex dynamic cellular organization in the hypothalamus is critical for understanding its role in coordinating fundamental body functions. Over the past decade, single-cell and spatial omics technologies have significantly evolved, overcoming initial technical challenges in capturing and analyzing individual cells. These high-throughput omics technologies now offer a remarkable opportunity to comprehend the complex spatiotemporal patterns of transcriptional diversity and cell-type characteristics across the entire hypothalamus. Current single-cell and single-nucleus RNA sequencing methods comprehensively quantify gene expression by exploring distinct phenotypes across various subregions of the hypothalamus. However, single-cell/single-nucleus RNA sequencing requires isolating the cell/nuclei from the tissue, potentially resulting in the loss of spatial information concerning neuronal networks. Spatial transcriptomics methods, by bypassing the cell dissociation, can elucidate the intricate spatial organization of neural networks through their imaging and sequencing technologies. In this review, we highlight the applicative value of single-cell and spatial transcriptomics in exploring the complex molecular-genetic diversity of hypothalamic cell types, driven by recent high-throughput achievements.
Collapse
Affiliation(s)
- Muhammad Junaid
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| | - Eun Jeong Lee
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| |
Collapse
|
2
|
Akter M, Fu Z, Zheng X, Iqbal Z, Zhang N, Karim A, Li Y. Astrocytic GPCR signaling in the anterior cingulate cortex modulates decision making in rats. OXFORD OPEN NEUROSCIENCE 2024; 3:kvae010. [PMID: 38915791 PMCID: PMC11194462 DOI: 10.1093/oons/kvae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024]
Abstract
Decision making is a process of selecting a course of action by assessing the worth or value of the potential consequences. Rat Gambling Task (RGT) is a well-established behavioral paradigm that allows for assessment of the decision-making performance of rats. Astrocytes are emerging as key players in modulating cognitive functions. Using repeated RGTs with short intersession time intervals (48 h), the current study demonstrates that Gi pathway activation of astrocytes in the anterior cingulate cortex (ACC) leads to impaired decision-making in consistently good decision-making rats. On the other hand, ACC astrocytic Gq pathway activation improves decision-making in a subset of rats who are not consistently good decision-makers. Furthermore, we show that astrocytic Gq activation is associated with an increase in the L-lactate level in the extracellular fluid of the ACC. Together, these results expand our knowledge of the role of astrocytic GPCR signaling in modulating cognitive functions.
Collapse
Affiliation(s)
- Mastura Akter
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Zhongqi Fu
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Xianlin Zheng
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Zafar Iqbal
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 15 Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong, SAR, China
- Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 17W, Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong, SAR, China
| | - Na Zhang
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Anwarul Karim
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| | - Ying Li
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 15 Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong, SAR, China
- Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 17W, Science Park West Avenue, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong, SAR, China
- Centre for Biosystems, Neuroscience, and Nanotechnology, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR, China
| |
Collapse
|
3
|
Iguchi Y, Fukabori R, Kato S, Takahashi K, Eifuku S, Maejima Y, Shimomura K, Mizuma H, Mawatari A, Doi H, Cui Y, Onoe H, Hikishima K, Osanai M, Nishijo T, Momiyama T, Benton R, Kobayashi K. Chemogenetic activation of mammalian brain neurons expressing insect Ionotropic Receptors by systemic ligand precursor administration. Commun Biol 2024; 7:547. [PMID: 38714803 PMCID: PMC11076466 DOI: 10.1038/s42003-024-06223-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/22/2024] [Indexed: 05/10/2024] Open
Abstract
Chemogenetic approaches employing ligand-gated ion channels are advantageous regarding manipulation of target neuronal population functions independently of endogenous second messenger pathways. Among them, Ionotropic Receptor (IR)-mediated neuronal activation (IRNA) allows stimulation of mammalian neurons that heterologously express members of the insect chemosensory IR repertoire in response to their cognate ligands. In the original protocol, phenylacetic acid, a ligand of the IR84a/IR8a complex, was locally injected into a brain region due to its low permeability of the blood-brain barrier. To circumvent this invasive injection, we sought to develop a strategy of peripheral administration with a precursor of phenylacetic acid, phenylacetic acid methyl ester, which is efficiently transferred into the brain and converted to the mature ligand by endogenous esterase activities. This strategy was validated by electrophysiological, biochemical, brain-imaging, and behavioral analyses, demonstrating high utility of systemic IRNA technology in the remote activation of target neurons in the brain.
Collapse
Affiliation(s)
- Yoshio Iguchi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Ryoji Fukabori
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Kazumi Takahashi
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Satoshi Eifuku
- Department of Systems Neuroscience, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Hiroshi Mizuma
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Aya Mawatari
- Laboratory for Labeling Chemistry, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Hisashi Doi
- Laboratory for Labeling Chemistry, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
- Research, Institute for Drug Discovery Science, Collaborative Creation Research Center, Organization for Research Promotion, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, 599-8531, Japan
| | - Yilong Cui
- Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Hirotaka Onoe
- Human Brain Research Center, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-Cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Keigo Hikishima
- Medical Devices Research Group, Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-2-1 Namiki, Tsukuba, 305-8564, Japan
| | - Makoto Osanai
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, 565-0871, Japan
| | - Takuma Nishijo
- Department of Pharmacology, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Tokyo, 105-8461, Japan
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya-cho, Kasugai, 480-0392, Japan
| | - Toshihiko Momiyama
- Department of Pharmacology, Jikei University School of Medicine, 3-25-8 Nishi-shinbashi, Tokyo, 105-8461, Japan
| | - Richard Benton
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima, 960-1295, Japan.
| |
Collapse
|
4
|
Kc E, Islam J, Kim HK, Park YS. GFAP-NpHR mediated optogenetic inhibition of trigeminal nucleus caudalis attenuates hypersensitive behaviors and thalamic discharge attributed to infraorbital nerve constriction injury. J Headache Pain 2023; 24:137. [PMID: 37821818 PMCID: PMC10566148 DOI: 10.1186/s10194-023-01669-z] [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: 08/09/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023] Open
Abstract
The significance of hyperactive astrocytes in neuropathic pain is crucial. However, the association between medullary astrocytes and trigeminal neuralgia (TN)-related pain processing is unclear. Here, we examined how optogenetic inhibition of medullary astrocytes in the trigeminal nucleus caudalis (TNC) regulates pain hypersensitivity in an infraorbital nerve (ION) constricted TN model. We used adult Sprague Dawley rats subjected to infraorbital nerve (ION) constriction to mimic TN symptoms, with naive and sham rats serving as controls. For in vivo optogenetic manipulations, rats stereotaxically received AAV8-GFAP-eNpHR3.0-mCherry or AAV8-GFAP-mCherry at the trigeminal nucleus caudalis (TNC). Open field, von Frey, air puff, and acetone tests measured pain behavioral flexibility. In vivo thalamic recordings were obtained simultaneously with optogenetic manipulation in the TNC. Orofacial hyperalgesia and thalamic hyperexcitability were both accompanied by medullary astrocyte hyperactivity, marked by upregulated GFAP. The yellow laser-driven inhibition of TNC astrocytes markedly improved behavioral responses and regulated thalamic neuronal responses. Halorhodopsin-mediated inhibition in medullary astrocytes may modify the nociceptive input transmitted through the trigeminothalamic tract and pain perception. Taken together, these findings imply that this subpopulation in the TNC and its thalamic connections play a significant role in regulating the trigeminal pain circuitry, which might aid in the identification of new therapeutic measures in TN management.
Collapse
Affiliation(s)
- Elina Kc
- Program in Neuroscience, Department of Medicine, College of Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Jaisan Islam
- Program in Neuroscience, Department of Medicine, College of Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Hyong Kyu Kim
- Department of Medicine and Microbiology, College of Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Young Seok Park
- Program in Neuroscience, Department of Medicine, College of Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea.
- Department of Neurosurgery, Chungbuk National University Hospital, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
5
|
Hyung S, Park JH, Jung K. Application of optogenetic glial cells to neuron-glial communication. Front Cell Neurosci 2023; 17:1249043. [PMID: 37868193 PMCID: PMC10585272 DOI: 10.3389/fncel.2023.1249043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/15/2023] [Indexed: 10/24/2023] Open
Abstract
Optogenetic techniques combine optics and genetics to enable cell-specific targeting and precise spatiotemporal control of excitable cells, and they are increasingly being employed. One of the most significant advantages of the optogenetic approach is that it allows for the modulation of nearby cells or circuits with millisecond precision, enabling researchers to gain a better understanding of the complex nervous system. Furthermore, optogenetic neuron activation permits the regulation of information processing in the brain, including synaptic activity and transmission, and also promotes nerve structure development. However, the optimal conditions remain unclear, and further research is required to identify the types of cells that can most effectively and precisely control nerve function. Recent studies have described optogenetic glial manipulation for coordinating the reciprocal communication between neurons and glia. Optogenetically stimulated glial cells can modulate information processing in the central nervous system and provide structural support for nerve fibers in the peripheral nervous system. These advances promote the effective use of optogenetics, although further experiments are needed. This review describes the critical role of glial cells in the nervous system and reviews the optogenetic applications of several types of glial cells, as well as their significance in neuron-glia interactions. Together, it briefly discusses the therapeutic potential and feasibility of optogenetics.
Collapse
Affiliation(s)
- Sujin Hyung
- Precision Medicine Research Institute, Samsung Medical Center, Seoul, Republic of Korea
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji-Hye Park
- Graduate School of Cancer Science and Policy, Cancer Biomedical Science, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Kyuhwan Jung
- DAWINBIO Inc., Hanam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
6
|
Brazhe A, Verisokin A, Verveyko D, Postnov D. Astrocytes: new evidence, new models, new roles. Biophys Rev 2023; 15:1303-1333. [PMID: 37975000 PMCID: PMC10643736 DOI: 10.1007/s12551-023-01145-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/08/2023] [Indexed: 11/19/2023] Open
Abstract
Astrocytes have been in the limelight of active research for about 3 decades now. Over this period, ideas about their function and role in the nervous system have evolved from simple assistance in energy supply and homeostasis maintenance to a complex informational and metabolic hub that integrates data on local neuronal activity, sensory and arousal context, and orchestrates many crucial processes in the brain. Rapid progress in experimental techniques and data analysis produces a growing body of data, which can be used as a foundation for formulation of new hypotheses, building new refined mathematical models, and ultimately should lead to a new level of understanding of the contribution of astrocytes to the cognitive tasks performed by the brain. Here, we highlight recent progress in astrocyte research, which we believe expands our understanding of how low-level signaling at a cellular level builds up to processes at the level of the whole brain and animal behavior. We start our review with revisiting data on the role of noradrenaline-mediated astrocytic signaling in locomotion, arousal, sensory integration, memory, and sleep. We then briefly review astrocyte contribution to the regulation of cerebral blood flow regulation, which is followed by a discussion of biophysical mechanisms underlying astrocyte effects on different brain processes. The experimental section is closed by an overview of recent experimental techniques available for modulation and visualization of astrocyte dynamics. We then evaluate how the new data can be potentially incorporated into the new mathematical models or where and how it already has been done. Finally, we discuss an interesting prospect that astrocytes may be key players in important processes such as the switching between sleep and wakefulness and the removal of toxic metabolites from the brain milieu.
Collapse
Affiliation(s)
- Alexey Brazhe
- Department of Biophysics, Biological Faculty, Lomonosov Moscow State University, Leninskie Gory, 1/24, Moscow, 119234 Russia
- Department of Molecular Neurobiology, Institute of Bioorganic Chemistry RAS, GSP-7, Miklukho-Maklay Str., 16/10, Moscow, 117997 Russia
| | - Andrey Verisokin
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, Kursk, 305000 Russia
| | - Darya Verveyko
- Department of Theoretical Physics, Kursk State University, Radishcheva st., 33, Kursk, 305000 Russia
| | - Dmitry Postnov
- Department of Optics and Biophotonics, Saratov State University, Astrakhanskaya st., 83, Saratov, 410012 Russia
| |
Collapse
|
7
|
Pereira MJ, Ayana R, Holt MG, Arckens L. Chemogenetic manipulation of astrocyte activity at the synapse- a gateway to manage brain disease. Front Cell Dev Biol 2023; 11:1193130. [PMID: 37534103 PMCID: PMC10393042 DOI: 10.3389/fcell.2023.1193130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 08/04/2023] Open
Abstract
Astrocytes are the major glial cell type in the central nervous system (CNS). Initially regarded as supportive cells, it is now recognized that this highly heterogeneous cell population is an indispensable modulator of brain development and function. Astrocytes secrete neuroactive molecules that regulate synapse formation and maturation. They also express hundreds of G protein-coupled receptors (GPCRs) that, once activated by neurotransmitters, trigger intracellular signalling pathways that can trigger the release of gliotransmitters which, in turn, modulate synaptic transmission and neuroplasticity. Considering this, it is not surprising that astrocytic dysfunction, leading to synaptic impairment, is consistently described as a factor in brain diseases, whether they emerge early or late in life due to genetic or environmental factors. Here, we provide an overview of the literature showing that activation of genetically engineered GPCRs, known as Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), to specifically modulate astrocyte activity partially mimics endogenous signalling pathways in astrocytes and improves neuronal function and behavior in normal animals and disease models. Therefore, we propose that expressing these genetically engineered GPCRs in astrocytes could be a promising strategy to explore (new) signalling pathways which can be used to manage brain disorders. The precise molecular, functional and behavioral effects of this type of manipulation, however, differ depending on the DREADD receptor used, targeted brain region and timing of the intervention, between healthy and disease conditions. This is likely a reflection of regional and disease/disease progression-associated astrocyte heterogeneity. Therefore, a thorough investigation of the effects of such astrocyte manipulation(s) must be conducted considering the specific cellular and molecular environment characteristic of each disease and disease stage before this has therapeutic applicability.
Collapse
Affiliation(s)
- Maria João Pereira
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Rajagopal Ayana
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Matthew G. Holt
- Instituto de Investigação e Inovação em Saúde (i3S), Laboratory of Synapse Biology, Universidade do Porto, Porto, Portugal
| | - Lutgarde Arckens
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
8
|
Kawano T, Zhou J, Anwar S, Salah H, Dayal AH, Ishikawa Y, Boetel K, Takahashi T, Sharma K, Inoue M. T cell infiltration into the brain triggers pulmonary dysfunction in murine Cryptococcus-associated IRIS. Nat Commun 2023; 14:3831. [PMID: 37380639 DOI: 10.1038/s41467-023-39518-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 06/16/2023] [Indexed: 06/30/2023] Open
Abstract
Cryptococcus-associated immune reconstitution inflammatory syndrome (C-IRIS) is a condition frequently occurring in immunocompromised patients receiving antiretroviral therapy. C-IRIS patients exhibit many critical symptoms, including pulmonary distress, potentially complicating the progression and recovery from this condition. Here, utilizing our previously established mouse model of unmasking C-IRIS (CnH99 preinfection and adoptive transfer of CD4+ T cells), we demonstrated that pulmonary dysfunction associated with the C-IRIS condition in mice could be attributed to the infiltration of CD4+ T cells into the brain via the CCL8-CCR5 axis, which triggers the nucleus tractus solitarius (NTS) neuronal damage and neuronal disconnection via upregulated ephrin B3 and semaphorin 6B in CD4+ T cells. Our findings provide unique insight into the mechanism behind pulmonary dysfunction in C-IRIS and nominate potential therapeutic targets for treatment.
Collapse
Affiliation(s)
- Tasuku Kawano
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima Aoba-Ku, Sendai, Miyagi, 981-8558, Japan
| | - Jinyan Zhou
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- Neuroscience Program, The University of Illinois at Urbana-Champaign, 405 North Matthews Avenue, Urbana, IL, 61801, USA
| | - Shehata Anwar
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University (BSU), Beni-Suef, 62511, Egypt
| | - Haneen Salah
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- School of Molecular and Cell Biology, The University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Andrea H Dayal
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- School of Molecular and Cell Biology, The University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Yuzuki Ishikawa
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- School of Molecular and Cell Biology, The University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Katelyn Boetel
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA
- School of Molecular and Cell Biology, The University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - Tomoko Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima Aoba-Ku, Sendai, Miyagi, 981-8558, Japan
| | - Kamal Sharma
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, 808 S. Wood Street, Chicago, IL, 60612, USA
| | - Makoto Inoue
- Department of Comparative Biosciences, The University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL, 61802, USA.
- Neuroscience Program, The University of Illinois at Urbana-Champaign, 405 North Matthews Avenue, Urbana, IL, 61801, USA.
- Beckman Institute for Advanced Science and Technology, 405 North Matthews Avenue, Urbana, IL, 61801, USA.
| |
Collapse
|
9
|
Ahn SH, Suh JS, Lim GH, Kim TJ. The Potential Effects of Light Irradiance in Glaucoma and Photobiomodulation Therapy. Bioengineering (Basel) 2023; 10:bioengineering10020223. [PMID: 36829717 PMCID: PMC9952036 DOI: 10.3390/bioengineering10020223] [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: 12/13/2022] [Revised: 01/29/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Human vision is mediated by the retina, one of the most critical tissues in the central nervous system. Glaucoma is a complex retinal disease attributed to environmental, genetic, and stochastic factors, all of which contribute to its pathogenesis. Historically, glaucoma had been thought of primarily as a disease of the elderly; however, it is now becoming more problematic as the incidence rate increases among young individuals. In recent years, excessive light exposure has been suggested as contributing to the rise in glaucoma among the younger generation. Blue light induces mitochondrial apoptosis in retinal ganglion cells, causing optic damage; red light increases cytochrome c oxidase activity in the electron transport system, reducing inflammation and increasing antioxidant reactions to promote cell regeneration. In conclusion, the minimization of blue light exposure and the general application of red light treatment strategies are anticipated to show synergistic effects with existing treatments for retinal disease and glaucoma and should be considered a necessary prospect for the future. This review introduces the recent studies that support the relationship between light exposure and the onset of glaucoma and discusses new treatments, such as photobiomodulation therapy.
Collapse
Affiliation(s)
- Sang-Hyun Ahn
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
| | - Jung-Soo Suh
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
| | - Gah-Hyun Lim
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
- Institute of Systems Biology, Pusan National University, Pusan 46241, Republic of Korea
- Correspondence: (G.-H.L.); (T.-J.K.); Tel.: +82-51-510-2261 (T.-J.K.)
| | - Tae-Jin Kim
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Pusan 46241, Republic of Korea
- Institute of Systems Biology, Pusan National University, Pusan 46241, Republic of Korea
- Correspondence: (G.-H.L.); (T.-J.K.); Tel.: +82-51-510-2261 (T.-J.K.)
| |
Collapse
|
10
|
Clyburn C, Carson KE, Smith CR, Travagli RA, Browning KN. Brainstem astrocytes control homeostatic regulation of caloric intake. J Physiol 2023; 601:801-829. [PMID: 36696965 PMCID: PMC10026361 DOI: 10.1113/jp283566] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 12/08/2022] [Indexed: 01/27/2023] Open
Abstract
Prolonged high-fat diet (HFD) exposure is associated with hyperphagia, excess caloric intake and weight gain. After initial exposure to a HFD, a brief (24-48 h) period of hyperphagia is followed by the regulation of caloric intake and restoration of energy balance within an acute (3-5 day) period. Previous studies have demonstrated this occurs via a vagally mediated signalling cascade that increases glutamatergic transmission via activation of NMDA receptors located on gastric-projecting neurons of the dorsal motor nucleus of the vagus (DMV). The present study used electrophysiological recordings from thin brainstem slice preparations, in vivo recordings of gastric motility and tone, measurement of gastric emptying rates, and food intake studies to investigate the hypothesis that activation of brainstem astrocytes in response to acute HFD exposure is responsible for the increased glutamatergic drive to DMV neurons and the restoration of caloric balance. Pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV excitability, dysregulated gastric tone and motility, attenuated the homeostatic delay in gastric emptying, and prevented the decrease in food intake that is observed during the period of energy regulation following initial exposure to HFD. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome. KEY POINTS: Initial exposure to a high fat diet is associated with a brief period of hyperphagia before caloric intake and energy balance is restored. This period of homeostatic regulation is associated with a vagally mediated signalling cascade that increases glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons via activation of synaptic NMDA receptors. The present study demonstrates that pharmacological and chemogenetic inhibition of brainstem astrocytes reduced glutamatergic signalling and DMV neuronal excitability, dysregulated gastric motility and tone and emptying, and prevented the regulation of food intake following high-fat diet exposure. Astrocyte regulation of glutamatergic transmission to DMV neurons appears to involve release of the gliotransmitters glutamate and ATP. Understanding the mechanisms involved in caloric regulation may provide critical insights into energy balance as well as into the hyperphagia that develops as these mechanisms are overcome.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
- Current position: Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, 97056
| | - Kaitlin E. Carson
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| | - Caleb R. Smith
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| | - R. Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
- Current position: Neurobiology Research, Newport, NC 28570
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA
| |
Collapse
|
11
|
Gorina YV, Salmina AB, Erofeev AI, Gerasimov EI, Bolshakova AV, Balaban PM, Bezprozvanny IB, Vlasova OL. Astrocyte Activation Markers. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:851-870. [PMID: 36180985 DOI: 10.1134/s0006297922090012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 06/16/2023]
Abstract
Astrocytes are the most common type of glial cells that provide homeostasis and protection of the central nervous system. Important specific characteristic of astrocytes is manifestation of morphological heterogeneity, which is directly dependent on localization in a particular area of the brain. Astrocytes can integrate into neural networks and keep neurons active in various areas of the brain. Moreover, astrocytes express a variety of receptors, channels, and membrane transporters, which underlie their peculiar metabolic activity, and, hence, determine plasticity of the central nervous system during development and aging. Such complex structural and functional organization of astrocytes requires the use of modern methods for their identification and analysis. Considering the important fact that determining the most appropriate marker for polymorphic and multiple subgroups of astrocytes is of decisive importance for studying their multifunctionality, this review presents markers, modern imaging techniques, and identification of astrocytes, which comprise a valuable resource for studying structural and functional properties of astrocytes, as well as facilitate better understanding of the extent to which astrocytes contribute to neuronal activity.
Collapse
Affiliation(s)
- Yana V Gorina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 194091, Russia.
- Research Institute of Molecular Medicine and Pathobiochemistry, Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
| | - Alla B Salmina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 194091, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, 660022, Russia
- Laboratory of Neurobiology and Tissue Engineering, Brain Institute, Research Center of Neurology, Moscow, 105064, Russia
| | - Alexander I Erofeev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 194091, Russia
| | - Evgeniy I Gerasimov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 194091, Russia
| | - Anastasia V Bolshakova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 194091, Russia
| | - Pavel M Balaban
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 194091, Russia
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity, Moscow, 117485, Russia
| | - Ilya B Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 194091, Russia
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Olga L Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 194091, Russia
| |
Collapse
|
12
|
Choi IS, Kim JH, Jeong JY, Lee MG, Suk K, Jang IS. Astrocyte-derived adenosine excites sleep-promoting neurons in the ventrolateral preoptic nucleus: Astrocyte-neuron interactions in the regulation of sleep. Glia 2022; 70:1864-1885. [PMID: 35638268 DOI: 10.1002/glia.24225] [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: 02/07/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/10/2022]
Abstract
Although ATP and/or adenosine derived from astrocytes are known to regulate sleep, the precise mechanisms underlying the somnogenic effects of ATP and adenosine remain unclear. We selectively expressed channelrhodopsin-2 (ChR2), a light-sensitive ion channel, in astrocytes within the ventrolateral preoptic nucleus (VLPO), which is an essential brain nucleus involved in sleep promotion. We then examined the effects of photostimulation of astrocytic ChR2 on neuronal excitability using whole-cell patch-clamp recordings in two functionally distinct types of VLPO neurons: sleep-promoting GABAergic projection neurons and non-sleep-promoting local GABAergic neurons. Optogenetic stimulation of VLPO astrocytes demonstrated opposite outcomes in the two types of VLPO neurons. It led to the inhibition of non-sleep-promoting neurons and excitation of sleep-promoting neurons. These responses were attenuated by blocking of either adenosine A1 receptors or tissue-nonspecific alkaline phosphatase (TNAP). In contrast, exogenous adenosine decreased the excitability of both VLPO neuron populations. Moreover, TNAP was expressed in galanin-negative VLPO neurons, but not in galanin-positive sleep-promoting projection neurons. Taken together, these results suggest that astrocyte-derived ATP is converted into adenosine by TNAP in non-sleep-promoting neurons. In turn, adenosine decreases the excitability of local GABAergic neurons, thereby increasing the excitability of sleep-promoting GABAergic projection neurons. We propose a novel mechanism involving astrocyte-neuron interactions in sleep regulation, wherein endogenous adenosine derived from astrocytes excites sleep-promoting VLPO neurons, and thus decreases neuronal excitability in arousal-related areas of the brain.
Collapse
Affiliation(s)
- In-Sun Choi
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Jae-Hong Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Ji-Young Jeong
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Maan-Gee Lee
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea.,Brain Science & Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, South Korea.,Brain Science & Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Il-Sung Jang
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, South Korea.,Brain Science & Engineering Institute, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
13
|
Miura Y, Senoo A, Doura T, Kiyonaka S. Chemogenetics of cell surface receptors: beyond genetic and pharmacological approaches. RSC Chem Biol 2022; 3:269-287. [PMID: 35359495 PMCID: PMC8905536 DOI: 10.1039/d1cb00195g] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/20/2022] [Indexed: 11/29/2022] Open
Abstract
Cell surface receptors transmit extracellular information into cells. Spatiotemporal regulation of receptor signaling is crucial for cellular functions, and dysregulation of signaling causes various diseases. Thus, it is highly desired to control receptor functions with high spatial and/or temporal resolution. Conventionally, genetic engineering or chemical ligands have been used to control receptor functions in cells. As the alternative, chemogenetics has been proposed, in which target proteins are genetically engineered to interact with a designed chemical partner with high selectivity. The engineered receptor dissects the function of one receptor member among a highly homologous receptor family in a cell-specific manner. Notably, some chemogenetic strategies have been used to reveal the receptor signaling of target cells in living animals. In this review, we summarize the developing chemogenetic methods of transmembrane receptors for cell-specific regulation of receptor signaling. We also discuss the prospects of chemogenetics for clinical applications.
Collapse
Affiliation(s)
- Yuta Miura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| | - Akinobu Senoo
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| | - Tomohiro Doura
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| | - Shigeki Kiyonaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Nagoya 464-8603 Japan
| |
Collapse
|
14
|
Lyon KA, Allen NJ. From Synapses to Circuits, Astrocytes Regulate Behavior. Front Neural Circuits 2022; 15:786293. [PMID: 35069124 PMCID: PMC8772456 DOI: 10.3389/fncir.2021.786293] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/05/2021] [Indexed: 12/21/2022] Open
Abstract
Astrocytes are non-neuronal cells that regulate synapses, neuronal circuits, and behavior. Astrocytes ensheath neuronal synapses to form the tripartite synapse where astrocytes influence synapse formation, function, and plasticity. Beyond the synapse, recent research has revealed that astrocyte influences on the nervous system extend to the modulation of neuronal circuitry and behavior. Here we review recent findings on the active role of astrocytes in behavioral modulation with a focus on in vivo studies, primarily in mice. Using tools to acutely manipulate astrocytes, such as optogenetics or chemogenetics, studies reviewed here have demonstrated a causal role for astrocytes in sleep, memory, sensorimotor behaviors, feeding, fear, anxiety, and cognitive processes like attention and behavioral flexibility. Current tools and future directions for astrocyte-specific manipulation, including methods for probing astrocyte heterogeneity, are discussed. Understanding the contribution of astrocytes to neuronal circuit activity and organismal behavior will be critical toward understanding how nervous system function gives rise to behavior.
Collapse
Affiliation(s)
- Krissy A Lyon
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Nicola J Allen
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
15
|
Novel Approaches Used to Examine and Control Neurogenesis in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22179608. [PMID: 34502516 PMCID: PMC8431772 DOI: 10.3390/ijms22179608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/16/2022] Open
Abstract
Neurogenesis is a key mechanism of brain development and plasticity, which is impaired in chronic neurodegeneration, including Parkinson’s disease. The accumulation of aberrant α-synuclein is one of the features of PD. Being secreted, this protein produces a prominent neurotoxic effect, alters synaptic plasticity, deregulates intercellular communication, and supports the development of neuroinflammation, thereby providing propagation of pathological events leading to the establishment of a PD-specific phenotype. Multidirectional and ambiguous effects of α-synuclein on adult neurogenesis suggest that impaired neurogenesis should be considered as a target for the prevention of cell loss and restoration of neurological functions. Thus, stimulation of endogenous neurogenesis or cell-replacement therapy with stem cell-derived differentiated neurons raises new hopes for the development of effective and safe technologies for treating PD neurodegeneration. Given the rapid development of optogenetics, it is not surprising that this method has already been repeatedly tested in manipulating neurogenesis in vivo and in vitro via targeting stem or progenitor cells. However, niche astrocytes could also serve as promising candidates for controlling neuronal differentiation and improving the functional integration of newly formed neurons within the brain tissue. In this review, we mainly focus on current approaches to assess neurogenesis and prospects in the application of optogenetic protocols to restore the neurogenesis in Parkinson’s disease.
Collapse
|
16
|
Malchow RP, Tchernookova BK, Choi JIV, Smith PJS, Kramer RH, Kreitzer MA. Review and Hypothesis: A Potential Common Link Between Glial Cells, Calcium Changes, Modulation of Synaptic Transmission, Spreading Depression, Migraine, and Epilepsy-H . Front Cell Neurosci 2021; 15:693095. [PMID: 34539347 PMCID: PMC8446203 DOI: 10.3389/fncel.2021.693095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/25/2021] [Indexed: 01/03/2023] Open
Abstract
There is significant evidence to support the notion that glial cells can modulate the strength of synaptic connections between nerve cells, and it has further been suggested that alterations in intracellular calcium are likely to play a key role in this process. However, the molecular mechanism(s) by which glial cells modulate neuronal signaling remains contentiously debated. Recent experiments have suggested that alterations in extracellular H+ efflux initiated by extracellular ATP may play a key role in the modulation of synaptic strength by radial glial cells in the retina and astrocytes throughout the brain. ATP-elicited alterations in H+ flux from radial glial cells were first detected from Müller cells enzymatically dissociated from the retina of tiger salamander using self-referencing H+-selective microelectrodes. The ATP-elicited alteration in H+ efflux was further found to be highly evolutionarily conserved, extending to Müller cells isolated from species as diverse as lamprey, skate, rat, mouse, monkey and human. More recently, self-referencing H+-selective electrodes have been used to detect ATP-elicited alterations in H+ efflux around individual mammalian astrocytes from the cortex and hippocampus. Tied to increases in intracellular calcium, these ATP-induced extracellular acidifications are well-positioned to be key mediators of synaptic modulation. In this article, we examine the evidence supporting H+ as a key modulator of neurotransmission, review data showing that extracellular ATP elicits an increase in H+ efflux from glial cells, and describe the potential signal transduction pathways involved in glial cell-mediated H+ efflux. We then examine the potential role that extracellular H+ released by glia might play in regulating synaptic transmission within the vertebrate retina, and then expand the focus to discuss potential roles in spreading depression, migraine, epilepsy, and alterations in brain rhythms, and suggest that alterations in extracellular H+ may be a unifying feature linking these disparate phenomena.
Collapse
Affiliation(s)
- Robert Paul Malchow
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Boriana K. Tchernookova
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Ji-in Vivien Choi
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States
- Stritch School of Medicine, Loyola University, Maywood, IL, United States
| | - Peter J. S. Smith
- Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton, United Kingdom
- Bell Center, Marine Biological Laboratory, Woods Hole, MA, United States
| | - Richard H. Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Matthew A. Kreitzer
- Department of Biology, Indiana Wesleyan University, Marion, IN, United States
| |
Collapse
|
17
|
Astrocytes in the Ventromedial Hypothalamus Involve Chronic Stress-Induced Anxiety and Bone Loss in Mice. Neural Plast 2021; 2021:7806370. [PMID: 34306063 PMCID: PMC8282369 DOI: 10.1155/2021/7806370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/22/2021] [Indexed: 11/18/2022] Open
Abstract
Chronic stress is one of the main risk factors of bone loss. While the neurons and neural circuits of the ventromedial hypothalamus (VMH) mediate bone loss induced by chronic stress, the detailed intrinsic mechanisms within the VMH nucleus still need to be explored. Astrocytes in brain regions play important roles in the regulation of metabolism and anxiety-like behavior through interactions with surrounding neurons. However, whether astrocytes in the VMH affect neuronal activity and therefore regulate chronic stress-induced anxiety and bone loss remain elusive. In this study, we found that VMH astrocytes were activated during chronic stress-induced anxiety and bone loss. Pharmacogenetic activation of the Gi and Gq pathways in VMH astrocytes reduced and increased the levels of anxiety and bone loss, respectively. Furthermore, activation of VMH astrocytes by optogenetics induced depolarization in neighboring steroidogenic factor-1 (SF-1) neurons, which was diminished by administration of N-methyl-D-aspartic acid (NMDA) receptor blocker but not by alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor blocker. These results suggest that there may be a functional "glial-neuron microcircuit" in VMH nuclei that mediates anxiety and bone loss induced by chronic stress. This study not only advances our understanding of glial cell function but also provides a potential intervention target for chronic stress-induced anxiety and bone loss therapy.
Collapse
|
18
|
McNeill J, Rudyk C, Hildebrand ME, Salmaso N. Ion Channels and Electrophysiological Properties of Astrocytes: Implications for Emergent Stimulation Technologies. Front Cell Neurosci 2021; 15:644126. [PMID: 34093129 PMCID: PMC8173131 DOI: 10.3389/fncel.2021.644126] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Astrocytes comprise a heterogeneous cell population characterized by distinct morphologies, protein expression and function. Unlike neurons, astrocytes do not generate action potentials, however, they are electrically dynamic cells with extensive electrophysiological heterogeneity and diversity. Astrocytes are hyperpolarized cells with low membrane resistance. They are heavily involved in the modulation of K+ and express an array of different voltage-dependent and voltage-independent channels to help with this ion regulation. In addition to these K+ channels, astrocytes also express several different types of Na+ channels; intracellular Na+ signaling in astrocytes has been linked to some of their functional properties. The physiological hallmark of astrocytes is their extensive intracellular Ca2+ signaling cascades, which vary at the regional, subregional, and cellular levels. In this review article, we highlight the physiological properties of astrocytes and the implications for their function and influence of network and synaptic activity. Furthermore, we discuss the implications of these differences in the context of optogenetic and DREADD experiments and consider whether these tools represent physiologically relevant techniques for the interrogation of astrocyte function.
Collapse
Affiliation(s)
- Jessica McNeill
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | | | | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
19
|
Bowles SN, Johnson CM. Inferences of glia-mediated control in Caenorhabditis elegans. J Neurosci Res 2021; 99:1191-1206. [PMID: 33559247 PMCID: PMC8005477 DOI: 10.1002/jnr.24803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/22/2022]
Abstract
Astrocytes modulate synaptic transmission; yet, it remains unclear how glia influence complex behaviors. Here, we explore the effects of Caenorhabditis elegans astrocyte-like cephalic glia (CEPglia ) and the glia-specific bHLH transcription factor HLH-17 on mating behavior and the defecation motor program (DMP). In C. elegans, male mating has been explicitly described through the male tail circuit and is characterized by coordination of multiple independent behaviors to ensure that copulation is achieved. Furthermore, the sex-specific male mating circuitry shares similar components with the DMP, which is complex and rhythmic, and requires a fixed sequence of behaviors to be activated periodically. We found that loss of CEPglia reduced persistence in executing mating behaviors and hindered copulation, while males that lacked HLH-17 demonstrated repetitive prodding behavior that increased the time spent in mating but did not hinder copulation. During the DMP, we found that posterior body wall contractions (pBocs) and enteric muscle contractions (EMCs) were differentially affected by loss of HLH-17 or CEPglia in males and hermaphrodites. pBocs and EMCs required HLH-17 activity in both sexes, whereas loss of CEPglia alone did not affect DMP in males. Our data suggest that CEPglia mediate complex behaviors by signaling to the GABAergic DVB neuron, and that HLH-17 activity influences those discrete steps within those behaviors. Collectively, these data provide evidence of glia as a link in cooperative regulation of complex and rhythmic behavior that, in C. elegans links circuitry in the head and the tail.
Collapse
Affiliation(s)
- Stephanie N. Bowles
- Department of Biology, Georgia State University, Atlanta, GA, 30303, United States
| | - Casonya M. Johnson
- Department of Biology, Georgia State University, Atlanta, GA, 30303, United States
- Department of Biology, James Madison University, Harrisonburg, VA, 22807
| |
Collapse
|
20
|
Choi JIV, Tchernookova BK, Kumar W, Kiedrowski L, Goeke C, Guizzetti M, Larson J, Kreitzer MA, Malchow RP. Extracellular ATP-Induced Alterations in Extracellular H + Fluxes From Cultured Cortical and Hippocampal Astrocytes. Front Cell Neurosci 2021; 15:640217. [PMID: 33994945 PMCID: PMC8120152 DOI: 10.3389/fncel.2021.640217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/19/2021] [Indexed: 12/18/2022] Open
Abstract
Small alterations in the level of extracellular H+ can profoundly alter neuronal activity throughout the nervous system. In this study, self-referencing H+-selective microelectrodes were used to examine extracellular H+ fluxes from individual astrocytes. Activation of astrocytes cultured from mouse hippocampus and rat cortex with extracellular ATP produced a pronounced increase in extracellular H+ flux. The ATP-elicited increase in H+ flux appeared to be independent of bicarbonate transport, as ATP increased H+ flux regardless of whether the primary extracellular pH buffer was 26 mM bicarbonate or 1 mM HEPES, and persisted when atmospheric levels of CO2 were replaced by oxygen. Adenosine failed to elicit any change in extracellular H+ fluxes, and ATP-mediated increases in H+ flux were inhibited by the P2 inhibitors suramin and PPADS suggesting direct activation of ATP receptors. Extracellular ATP also induced an intracellular rise in calcium in cultured astrocytes, and ATP-induced rises in both calcium and H+ efflux were significantly attenuated when calcium re-loading into the endoplasmic reticulum was inhibited by thapsigargin. Replacement of extracellular sodium with choline did not significantly reduce the size of the ATP-induced increases in H+ flux, and the increases in H+ flux were not significantly affected by addition of EIPA, suggesting little involvement of Na+/H+ exchangers in ATP-elicited increases in H+ flux. Given the high sensitivity of voltage-sensitive calcium channels on neurons to small changes in levels of free H+, we hypothesize that the ATP-mediated extrusion of H+ from astrocytes may play a key role in regulating signaling at synapses within the nervous system.
Collapse
Affiliation(s)
- Ji-In Vivien Choi
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States.,Stritch School of Medicine, Loyola University, Maywood, IL, United States
| | - Boriana K Tchernookova
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Wasan Kumar
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Lech Kiedrowski
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States.,Spot Cells LLC, Chicago, IL, United States
| | - Calla Goeke
- VA Portland Health Care System, Portland, OR, United States.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Marina Guizzetti
- VA Portland Health Care System, Portland, OR, United States.,Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - John Larson
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Matthew A Kreitzer
- Department of Biology, Indiana Wesleyan University, Marion, IN, United States
| | - Robert Paul Malchow
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States.,Department Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
21
|
Salmina AB, Gorina YV, Erofeev AI, Balaban PM, Bezprozvanny IB, Vlasova OL. Optogenetic and chemogenetic modulation of astroglial secretory phenotype. Rev Neurosci 2021; 32:459-479. [PMID: 33550788 DOI: 10.1515/revneuro-2020-0119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 11/28/2020] [Indexed: 12/20/2022]
Abstract
Astrocytes play a major role in brain function and alterations in astrocyte function that contribute to the pathogenesis of many brain disorders. The astrocytes are attractive cellular targets for neuroprotection and brain tissue regeneration. Development of novel approaches to monitor and to control astroglial function is of great importance for further progress in basic neurobiology and in clinical neurology, as well as psychiatry. Recently developed advanced optogenetic and chemogenetic techniques enable precise stimulation of astrocytes in vitro and in vivo, which can be achieved by the expression of light-sensitive channels and receptors, or by expression of receptors exclusively activated by designer drugs. Optogenetic stimulation of astrocytes leads to dramatic changes in intracellular calcium concentrations and causes the release of gliotransmitters. Optogenetic and chemogenetic protocols for astrocyte activation aid in extracting novel information regarding the function of brain's neurovascular unit. This review summarizes current data obtained by this approach and discusses a potential mechanistic connection between astrocyte stimulation and changes in brain physiology.
Collapse
Affiliation(s)
- Alla B Salmina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Yana V Gorina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Alexander I Erofeev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Pavel M Balaban
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Ilya B 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
| | - Olga L Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
22
|
Hwang SN, Lee JS, Seo K, Lee H. Astrocytic Regulation of Neural Circuits Underlying Behaviors. Cells 2021; 10:cells10020296. [PMID: 33535587 PMCID: PMC7912785 DOI: 10.3390/cells10020296] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
Astrocytes, characterized by a satellite-like morphology, are the most abundant type of glia in the central nervous system. Their main functions have been thought to be limited to providing homeostatic support for neurons, but recent studies have revealed that astrocytes actually actively interact with local neural circuits and play a crucial role in information processing and generating physiological and behavioral responses. Here, we review the emerging roles of astrocytes in many brain regions, particularly by focusing on intracellular changes in astrocytes and their interactions with neurons at the molecular and neural circuit levels.
Collapse
Affiliation(s)
- Sun-Nyoung Hwang
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
| | - Jae Seung Lee
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Korea; (J.S.L.); (K.S.)
| | - Kain Seo
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Korea; (J.S.L.); (K.S.)
| | - Hyosang Lee
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Korea; (J.S.L.); (K.S.)
- Korea Brain Research Institute (KBRI), Daegu 41062, Korea
- Correspondence: ; Tel.: +82-53-785-6147
| |
Collapse
|
23
|
Ouali Alami N, Tang L, Wiesner D, Commisso B, Bayer D, Weishaupt J, Dupuis L, Wong P, Baumann B, Wirth T, Boeckers TM, Yilmazer-Hanke D, Ludolph A, Roselli F. Multiplexed chemogenetics in astrocytes and motoneurons restore blood-spinal cord barrier in ALS. Life Sci Alliance 2020; 3:3/11/e201900571. [PMID: 32900826 PMCID: PMC7479971 DOI: 10.26508/lsa.201900571] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022] Open
Abstract
Chemogenetic motoneuron excitation and astrocyte GPCR-Gi signaling restore blood–spinal cord barrier, disrupted in four ALS mouse models, revealing its role in disease progression but not initiation. Blood–spinal cord barrier (BSCB) disruption is thought to contribute to motoneuron (MN) loss in amyotrophic lateral sclerosis (ALS). It is currently unclear whether impairment of the BSCB is the cause or consequence of MN dysfunction and whether its restoration may be directly beneficial. We revealed that SOD1G93A, FUSΔNLS, TDP43G298S, and Tbk1+/− ALS mouse models commonly shared alterations in the BSCB, unrelated to motoneuron loss. We exploit PSAM/PSEM chemogenetics in SOD1G93A mice to demonstrate that the BSCB is rescued by increased MN firing, whereas inactivation worsens it. Moreover, we use DREADD chemogenetics, alone or in multiplexed form, to show that activation of Gi signaling in astrocytes restores BSCB integrity, independently of MN firing, with no effect on MN disease markers and dissociating them from BSCB disruption. We show that astrocytic levels of the BSCB stabilizers Wnt7a and Wnt5a are decreased in SOD1G93A mice and strongly enhanced by Gi signaling, although further decreased by MN inactivation. Thus, we demonstrate that BSCB impairment follows MN dysfunction in ALS pathogenesis but can be reversed by Gi-induced expression of astrocytic Wnt5a/7a.
Collapse
Affiliation(s)
- Najwa Ouali Alami
- Department of Neurology, Ulm University, Ulm, Germany.,International Graduate School in Molecular Medicine Ulm, Ulm, Germany.,Department of Neurology, Clinical Neuroanatomy, Ulm University, Ulm, Germany
| | - Linyun Tang
- Department of Neurology, Ulm University, Ulm, Germany
| | - Diana Wiesner
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | | | - David Bayer
- Department of Neurology, Ulm University, Ulm, Germany.,CEMMA Graduate School, Ulm University, Ulm, Germany
| | | | - Luc Dupuis
- Inserm U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence; Université de Strasbourg, Faculté de Médecine, Strasbourg, France
| | - Phillip Wong
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Tobias M Boeckers
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany.,Department of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Albert Ludolph
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany .,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| |
Collapse
|
24
|
Li Y, Li L, Wu J, Zhu Z, Feng X, Qin L, Zhu Y, Sun L, Liu Y, Qiu Z, Duan S, Yu YQ. Activation of astrocytes in hippocampus decreases fear memory through adenosine A 1 receptors. eLife 2020; 9:57155. [PMID: 32869747 PMCID: PMC7505657 DOI: 10.7554/elife.57155] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022] Open
Abstract
Astrocytes respond to and regulate neuronal activity, yet their role in mammalian behavior remains incompletely understood. Especially unclear is whether, and if so how, astrocyte activity regulates contextual fear memory, the dysregulation of which leads to pathological fear-related disorders. We generated GFAP-ChR2-EYFP rats to allow the specific activation of astrocytes in vivo by optogenetics. We found that after memory acquisition within a temporal window, astrocyte activation disrupted memory consolidation and persistently decreased contextual but not cued fear memory accompanied by reduced fear-related anxiety behavior. In vivo microdialysis experiments showed astrocyte photoactivation increased extracellular ATP and adenosine concentrations. Intracerebral blockade of adenosine A1 receptors (A1Rs) reversed the attenuation of fear memory. Furthermore, intracerebral or intraperitoneal injection of A1R agonist mimicked the effects of astrocyte activation. Therefore, our findings provide a deeper understanding of the astrocyte-mediated regulation of fear memory and suggest a new and important therapeutic strategy against pathological fear-related disorders. Memory is the record of what we learn over time and is essential to our survival. But not all memories are helpful. Repeatedly recalling a traumatic event – such as an assault – can be harmful. About 1 in 3 people who experience severe trauma go on to develop post-traumatic stress disorder (PTSD), in which they re-live the traumatic event in the form of flashbacks and nightmares. Others develop panic disorder, phobias or depression. Preventing this chain of events is challenging because fear memories form rapidly and last a long time. Current treatments involve re-exposing individuals to the traumatic event. This could be real-life exposure in the case of a phobia. Or it could involve visualizing the event, in the case of PTSD. Controlled re-exposure can help individuals learn new coping strategies. But it does not erase the initial fear memory. A better approach might be to take advantage of the fact that new memories are unstable. To form a long-lasting memory trace, newly acquired information must go through a process called consolidation to stabilize it. This process takes place in an area of the brain called the hippocampus. If consolidation does not occur, new memory traces can fade away. Li, Li et al. now show that preventing consolidation in the rat brain stops the animals from forming lasting memories of a stressful event, namely a foot shock. In the study, the rats first learned to associate a foot shock with a tone. This training took place inside a specific chamber. After learning the association, the rats began to freeze – a sign of fear – whenever they entered the chamber. This happened even if the tone was not played. But Li, Li et al. showed that they could reduce this fear response by activating cells in the hippocampus known as astrocytes, shortly after the learning episode. Activating the astrocytes made them release a substance called adenosine. Molecules of adenosine then bound to and activated proteins called adenosine A1 receptors. Administering a drug that activated these receptors directly had the same effect as activating the astrocytes themselves. This suggests that drugs of this type could one day help patients with fear-related disorders such as PTSD and phobias. For this to become a reality, new studies must test different drugs and find the best ways of administering them. After testing in animal models, the next step will be preliminary clinical trials in people.
Collapse
Affiliation(s)
- Yulan Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Lixuan Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jintao Wu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Zhenggang Zhu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Xiang Feng
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Liming Qin
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Yuwei Zhu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Li Sun
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Yijun Liu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Zilong Qiu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.,Research Units for Emotion and Emotion Disorders, Chinese Academy of Medical Sciences, Hangzhou, China
| | - Yan-Qin Yu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Hao T, Du X, Yang S, Zhang Y, Liang F. Astrocytes-induced neuronal inhibition contributes to depressive-like behaviors during chronic stress. Life Sci 2020; 258:118099. [PMID: 32682917 DOI: 10.1016/j.lfs.2020.118099] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/01/2020] [Accepted: 07/10/2020] [Indexed: 01/09/2023]
Abstract
Although emerging evidence has highlighted the heterogeneities of astrocytes under physiological versus pathological conditions, little is known regarding these processes in different brain regions during stress. Thus, the present study established a mouse model of chronic social defeat stress (CSDS) and isolated astrocytes from the medial prefrontal cortex (mPFC) and hippocampus. The results revealed dramatic A1-specific (neurotoxic phenotype) astrocytic responses, depressive-like behaviors, and significant inhibition of neuronal activities in both the mPFC and hippocampus according to electrophysiological data. Subsequently, astrocytes in the mPFC and hippocampus of CSDS mice were suppressed and this reversed the astrocytic responses and rescued depressive-like behaviors. Furthermore, when astrocytes were activated in the mPFC and hippocampus in healthy mice, there was a non-specific phenotypic activation of astrocytes in the absence of depressive-like behaviors. Next, microglia were depleted and the mice subsequently performed in the CSDS model; this reduced astrocyte responses and restored depressive-like behaviors. On the other hand, when microglia were depleted but astrocytes were activated in CSDS mice, this abolished the restoration of microglia depletion-induced depressive-like behaviors. Taken together, these results indicate that neuronal inhibition by astrocytes in the mPFC and hippocampus contributed to depressive-like behaviors mediated by activated microglia. This study provides evidence regarding the interaction of microglia and astrocytes during stress and how that relationship can trigger depressive-like behaviors.
Collapse
Affiliation(s)
- Tianpao Hao
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaohong Du
- Department of geriatric medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shen Yang
- Departments of Neurology, Tai'an City Central Hospital, Tai'an 271000, China
| | - Yang Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Feiyu Liang
- Department of geriatric medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
26
|
Octeau JC, Gangwani MR, Allam SL, Tran D, Huang S, Hoang-Trong TM, Golshani P, Rumbell TH, Kozloski JR, Khakh BS. Transient, Consequential Increases in Extracellular Potassium Ions Accompany Channelrhodopsin2 Excitation. Cell Rep 2020; 27:2249-2261.e7. [PMID: 31116972 PMCID: PMC6582980 DOI: 10.1016/j.celrep.2019.04.078] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/04/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
Channelrhodopsin2 (ChR2) optogenetic excitation is widely used to study neurons, astrocytes, and circuits. Using complementary approaches in situ and in vivo, we found that ChR2 stimulation leads to significant transient elevation of extracellular potassium ions by ~5 mM. Such elevations were detected in ChR2-expressing mice, following local in vivo expression of ChR2(H134R) with adeno-associated viruses (AAVs), in different brain areas and when ChR2 was expressed in neurons or astrocytes. In particular, ChR2-mediated excitation of striatal astrocytes was sufficient to increase medium spiny neuron (MSN) excitability and immediate early gene expression. The effects on MSN excitability were recapitulated in silico with a computational MSN model and detected in vivo as increased action potential firing in awake, behaving mice. We show that transient, physiologically consequential increases in extracellular potassium ions accompany ChR2 optogenetic excitation. This coincidental effect may be important to consider during astrocyte studies employing ChR2 to interrogate neural circuits and animal behavior. Using multiple approaches, Octeau et al. discover that optogenetic excitation of ChR2-expressing cells leads to significant transient extracellular potassium ion elevations that increase neuronal excitability and immediate early gene expression in neurons following in vivo stimulation.
Collapse
Affiliation(s)
- J Christopher Octeau
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Mohitkumar R Gangwani
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Sushmita L Allam
- IBM T.J. Watson Research Center, P.O. Box 218, 1101 Kitchawan Road, Yorktown Heights, NY 10598, USA
| | - Duy Tran
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Shuhan Huang
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Tuan M Hoang-Trong
- IBM T.J. Watson Research Center, P.O. Box 218, 1101 Kitchawan Road, Yorktown Heights, NY 10598, USA
| | - Peyman Golshani
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Timothy H Rumbell
- IBM T.J. Watson Research Center, P.O. Box 218, 1101 Kitchawan Road, Yorktown Heights, NY 10598, USA
| | - James R Kozloski
- IBM T.J. Watson Research Center, P.O. Box 218, 1101 Kitchawan Road, Yorktown Heights, NY 10598, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
27
|
Chemogenetics a robust approach to pharmacology and gene therapy. Biochem Pharmacol 2020; 175:113889. [DOI: 10.1016/j.bcp.2020.113889] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
|
28
|
Wang YC, Galeffi F, Wang W, Li X, Lu L, Sheng H, Hoffmann U, Turner DA, Yang W. Chemogenetics-mediated acute inhibition of excitatory neuronal activity improves stroke outcome. Exp Neurol 2020; 326:113206. [PMID: 31962128 DOI: 10.1016/j.expneurol.2020.113206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/07/2020] [Accepted: 01/17/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND PURPOSE Ischemic stroke significantly perturbs neuronal homeostasis leading to a cascade of pathologic events causing brain damage. In this study, we assessed acute stroke outcome after chemogenetic inhibition of forebrain excitatory neuronal activity. METHODS We generated hM4Di-TG transgenic mice expressing the inhibitory hM4Di, a Designer Receptors Exclusively Activated by Designer Drugs (DREADD)-based chemogenetic receptor, in forebrain excitatory neurons. Clozapine-N-oxide (CNO) was used to activate hM4Di DREADD. Ischemic stroke was induced by transient occlusion of the middle cerebral artery. Neurologic function and infarct volumes were evaluated. Excitatory neuronal suppression in the hM4Di-TG mouse forebrain was assessed electrophysiologically in vitro and in vivo, based on evoked synaptic responses, and in vivo based on occurrence of potassium-induced cortical spreading depolarizations. RESULTS Detailed characterization of hM4Di-TG mice confirmed that evoked synaptic responses in both in vitro hippocampal slices and in vivo motor cortex were significantly reduced after CNO-mediated activation of the inhibitory hM4Di DREADD. Further, CNO treatment had no obvious effects on physiology and motor function in either control or hM4Di-TG mice. Importantly, hM4Di-TG mice treated with CNO at either 10 min before ischemia or 30 min after reperfusion exhibited significantly improved neurologic function and smaller infarct volumes compared to CNO-treated control mice. Mechanistically, we showed that potassium-induced cortical spreading depression episodes were inhibited, including frequency and duration of DC shift, in CNO-treated hM4Di-TG mice. CONCLUSIONS Our data demonstrate that acute inhibition of a subset of excitatory neurons after ischemic stroke can prevent brain injury and improve functional outcome. This study, together with the previous work in optogenetic neuronal modulation during the chronic phase of stroke, supports the notion that targeting neuronal activity is a promising strategy in stroke therapy.
Collapse
Affiliation(s)
- Ya-Chao Wang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | | | - Wei Wang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Department of Anesthesiology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, China
| | - Xuan Li
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Liping Lu
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Huaxin Sheng
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Ulrike Hoffmann
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Dennis A Turner
- Research and Surgery Services, Durham VAMC, Durham, NC, USA; Departments of Neurosurgery, Neurobiology and Biomedical Engineering, Duke University Medical Center, Durham, NC, USA
| | - Wei Yang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
29
|
Yang R, Dzowo YK, Wilson CE, Russell RL, Kidd GJ, Salcedo E, Lasher RS, Kinnamon JC, Finger TE. Three-dimensional reconstructions of mouse circumvallate taste buds using serial blockface scanning electron microscopy: I. Cell types and the apical region of the taste bud. J Comp Neurol 2019; 528:756-771. [PMID: 31587284 DOI: 10.1002/cne.24779] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/21/2019] [Accepted: 09/07/2019] [Indexed: 12/20/2022]
Abstract
Taste buds comprise four types of taste cells: three mature, elongate types, Types I-III; and basally situated, immature postmitotic type, Type IV cells. We employed serial blockface scanning electron microscopy to delineate the characteristics and interrelationships of the taste cells in the circumvallate papillae of adult mice. Type I cells have an indented, elongate nucleus with invaginations, folded plasma membrane, and multiple apical microvilli in the taste pore. Type I microvilli may be either restricted to the bottom of the pore or extend outward reaching midway up into the taste pore. Type II cells (aka receptor cells) possess a large round or oval nucleus, a single apical microvillus extending through the taste pore, and specialized "atypical" mitochondria at functional points of contact with nerve fibers. Type III cells (aka "synaptic cells") are elongate with an indented nucleus, possess a single, apical microvillus extending through the taste pore, and are characterized by a small accumulation of synaptic vesicles at points of contact with nerve fibers. About one-quarter of Type III cells also exhibit an atypical mitochondrion near the presynaptic vesicle clusters at the synapse. Type IV cells (nonproliferative "basal cells") have a nucleus in the lower quarter of the taste bud and a foot process extending to the basement membrane often contacting nerve processes along the way. In murine circumvallate taste buds, Type I cells represent just over 50% of the population, whereas Types II, III, and IV (basal cells) represent 19, 15, and 14%, respectively.
Collapse
Affiliation(s)
- Ruibiao Yang
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Denver, Colorado
| | - Yannick K Dzowo
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Denver, Colorado.,Modern Human Anatomy Program, University of Colorado School of Medicine, Denver, Colorado
| | - Courtney E Wilson
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Denver, Colorado
| | - Rae L Russell
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Denver, Colorado.,Modern Human Anatomy Program, University of Colorado School of Medicine, Denver, Colorado
| | - Grahame J Kidd
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,3D-Electron Microscopy, Renovo Neural Inc., Cleveland, Ohio
| | - Ernesto Salcedo
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Denver, Colorado.,Modern Human Anatomy Program, University of Colorado School of Medicine, Denver, Colorado
| | - Robert S Lasher
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Denver, Colorado
| | - John C Kinnamon
- Department of Biological Sciences, University of Denver, Denver, Colorado
| | - Thomas E Finger
- Department of Cell and Developmental Biology, Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Denver, Colorado.,Modern Human Anatomy Program, University of Colorado School of Medicine, Denver, Colorado
| |
Collapse
|
30
|
Biosensors for Epilepsy Management: State-of-Art and Future Aspects. SENSORS 2019; 19:s19071525. [PMID: 30925837 PMCID: PMC6480455 DOI: 10.3390/s19071525] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/24/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022]
Abstract
Epilepsy is a serious neurological disorder which affects every aspect of patients’ life, including added socio-economic burden. Unfortunately, only a few suppressive medicines are available, and a complete cure for the disease has not been found yet. Excluding the effectiveness of available therapies, the timely detection and monitoring of epilepsy are of utmost priority for early remediation and prevention. Inability to detect underlying epileptic signatures at early stage causes serious damage to the central nervous system (CNS) and irreversible detrimental variations in the organ system. Therefore, development of a multi-task solving novel smart biosensing systems is urgently required. The present review highlights advancements in state-of-art biosensing technology investigated for epilepsy diseases diagnostics and progression monitoring or both together. State of art epilepsy biosensors are composed of nano-enabled smart sensing platform integrated with micro/electronics and display. These diagnostics systems provide bio-information needed to understand disease progression and therapy optimization timely. The associated challenges related to the development of an efficient epilepsy biosensor and vision considering future prospects are also discussed in this report. This review will serve as a guide platform to scholars for understanding and planning of future research aiming to develop a smart bio-sensing system to detect and monitor epilepsy for point-of-care (PoC) applications.
Collapse
|
31
|
Mederos S, Hernández-Vivanco A, Ramírez-Franco J, Martín-Fernández M, Navarrete M, Yang A, Boyden ES, Perea G. Melanopsin for precise optogenetic activation of astrocyte-neuron networks. Glia 2019; 67:915-934. [PMID: 30632636 DOI: 10.1002/glia.23580] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022]
Abstract
Optogenetics has been widely expanded to enhance or suppress neuronal activity and it has been recently applied to glial cells. Here, we have used a new approach based on selective expression of melanopsin, a G-protein-coupled photopigment, in astrocytes to trigger Ca2+ signaling. Using the genetically encoded Ca2+ indicator GCaMP6f and two-photon imaging, we show that melanopsin is both competent to stimulate robust IP3-dependent Ca2+ signals in astrocyte fine processes, and to evoke an ATP/Adenosine-dependent transient boost of hippocampal excitatory synaptic transmission. Additionally, under low-frequency light stimulation conditions, melanopsin-transfected astrocytes can trigger long-term synaptic changes. In vivo, melanopsin-astrocyte activation enhances episodic-like memory, suggesting melanopsin as an optical tool that could recapitulate the wide range of regulatory actions of astrocytes on neuronal networks in behaving animals. These results describe a novel approach using melanopsin as a precise trigger for astrocytes that mimics their endogenous G-protein signaling pathways, and present melanopsin as a valuable optical tool for neuron-glia studies.
Collapse
Affiliation(s)
- Sara Mederos
- Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC, Madrid, Spain
| | | | - Jorge Ramírez-Franco
- Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC, Madrid, Spain
| | | | - Marta Navarrete
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Aimei Yang
- Media Lab, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Edward S Boyden
- Media Lab, Massachusetts Institute of Technology, Cambridge, Massachusetts.,McGovern Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Gertrudis Perea
- Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC, Madrid, Spain
| |
Collapse
|
32
|
Scofield MD. Exploring the Role of Astroglial Glutamate Release and Association With Synapses in Neuronal Function and Behavior. Biol Psychiatry 2018; 84:778-786. [PMID: 29258653 PMCID: PMC5948108 DOI: 10.1016/j.biopsych.2017.10.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/18/2017] [Accepted: 10/31/2017] [Indexed: 12/25/2022]
Abstract
Astrocytes are stellate cells whose appearance can resemble a pointed star, especially when visualizing glial fibrillary acidic protein, a canonical marker for astrocytes. Accordingly, there is a commonly made connection between the points of light that shine in the night sky and the diffuse and abundant cells that buffer ions and provide support for neurons. An exceptional amount of function has been attributed to, negated for, and potentially reaffirmed for these cells, especially regarding their ability to release neuroactive molecules and influence synaptic plasticity. This makes the precise role of astrocytes in tuning neural communication seem difficult to grasp. However, data from animal models of addiction demonstrate that a variety of drug-induced molecular adaptations responsible for relapse vulnerability take place in astrocyte systems that regulate glutamate uptake and release. These findings highlight astrocytes as a critical component of the neural systems responsible for addiction, serving as a key component of the plasticity responsible for relapse and drug seeking. Here I assemble recent findings that utilize genetic tools to selectively manipulate or measure flux of internal calcium in astrocytes, focusing on G protein-coupled receptor-mediated mobilization of calcium and the induction of glutamate release. Further, I compile evidence regarding astrocyte glutamate release as well as astrocyte association with synapses with respect to the impact of these cellular phenomena in shaping synaptic transmission. I also place these findings in the context of the previous studies of Scofield et al., who explored the role of astrocytes in the nucleus accumbens in the neural mechanisms underlying cocaine seeking.
Collapse
Affiliation(s)
- Michael D. Scofield
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, 29425 USA,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425 USA
| |
Collapse
|
33
|
Pál B. Involvement of extrasynaptic glutamate in physiological and pathophysiological changes of neuronal excitability. Cell Mol Life Sci 2018; 75:2917-2949. [PMID: 29766217 PMCID: PMC11105518 DOI: 10.1007/s00018-018-2837-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Glutamate is the most abundant neurotransmitter of the central nervous system, as the majority of neurons use glutamate as neurotransmitter. It is also well known that this neurotransmitter is not restricted to synaptic clefts, but found in the extrasynaptic regions as ambient glutamate. Extrasynaptic glutamate originates from spillover of synaptic release, as well as from astrocytes and microglia. Its concentration is magnitudes lower than in the synaptic cleft, but receptors responding to it have higher affinity for it. Extrasynaptic glutamate receptors can be found in neuronal somatodendritic location, on astroglia, oligodendrocytes or microglia. Activation of them leads to changes of neuronal excitability with different amplitude and kinetics. Extrasynaptic glutamate is taken up by neurons and astrocytes mostly via EAAT transporters, and astrocytes, in turn metabolize it to glutamine. Extrasynaptic glutamate is involved in several physiological phenomena of the central nervous system. It regulates neuronal excitability and synaptic strength by involving astroglia; contributing to learning and memory formation, neurosecretory and neuromodulatory mechanisms, as well as sleep homeostasis.The extrasynaptic glutamatergic system is affected in several brain pathologies related to excitotoxicity, neurodegeneration or neuroinflammation. Being present in dementias, neurodegenerative and neuropsychiatric diseases or tumor invasion in a seemingly uniform way, the system possibly provides a common component of their pathogenesis. Although parts of the system are extensively discussed by several recent reviews, in this review I attempt to summarize physiological actions of the extrasynaptic glutamate on neuronal excitability and provide a brief insight to its pathology for basic understanding of the topic.
Collapse
Affiliation(s)
- Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Nagyerdei krt 98, Debrecen, 4012, Hungary.
| |
Collapse
|
34
|
Cell reprogramming approaches in gene- and cell-based therapies for Parkinson's disease. J Control Release 2018; 286:114-124. [PMID: 30026082 DOI: 10.1016/j.jconrel.2018.07.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/26/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022]
Abstract
Degeneration of dopamine (DA) neurons in the substantia nigra pars compacta is the pathological hallmark of Parkinson's disease (PD). In PD multiple pathogenic mechanisms initiate and drive this neurodegenerative process, making the development of effective treatments challenging. To date, PD patients are primarily treated with dopaminergic drugs able to temporarily enhance DA levels, therefore relieving motor symptoms. However, the drawbacks of these therapies including the inability to alter disease progression are constantly supporting the search for alternative treatment approaches. Over the past years efforts have been put into the development of new therapeutic strategies based on the delivery of therapeutic genes using viral vectors or transplantation of DA neurons for cell-based DA replacement. Here, past achievements and recent advances in gene- and cell-based therapies for PD are outlined. We discuss how current gene and cell therapy strategies hold great promise for the treatment of PD and how the use of stem cells and recent developments in cellular reprogramming could contribute to open a new avenue in PD therapy.
Collapse
|
35
|
Rosso MPDO, Buchaim DV, Kawano N, Furlanette G, Pomini KT, Buchaim RL. Photobiomodulation Therapy (PBMT) in Peripheral Nerve Regeneration: A Systematic Review. Bioengineering (Basel) 2018; 5:bioengineering5020044. [PMID: 29890728 PMCID: PMC6027218 DOI: 10.3390/bioengineering5020044] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 12/18/2022] Open
Abstract
Photobiomodulation therapy (PBMT) has been investigated because of its intimate relationship with tissue recovery processes, such as on peripheral nerve damage. Based on the wide range of benefits that the PBMT has shown and its clinical relevance, the aim of this research was to carry out a systematic review of the last 10 years, ascertaining the influence of the PBMT in the regeneration of injured peripheral nerves. The search was performed in the PubMed/MEDLINE database with the combination of the keywords: low-level laser therapy AND nerve regeneration. Initially, 54 articles were obtained, 26 articles of which were chosen for the study according to the inclusion criteria. In the qualitative aspect, it was observed that PBMT was able to accelerate the process of nerve regeneration, presenting an increase in the number of myelinated fibers and a better lamellar organization of myelin sheath, besides improvement of electrophysiological function, immunoreactivity, high functionality rate, decrease of inflammation, pain, and the facilitation of neural regeneration, release of growth factors, increase of vascular network and collagen. It was concluded that PBMT has beneficial effects on the recovery of nerve lesions, especially when related to a faster regeneration and functional improvement, despite the variety of parameters.
Collapse
Affiliation(s)
- Marcelie Priscila de Oliveira Rosso
- Department of Biological Sciences (Anatomy), Bauru School of Dentistry, University of São Paulo (USP), Alameda Dr. Octávio Pinheiro Brisola 9-75, Vila Nova Cidade Universitária, Bauru, São Paulo CEP 17012-901, Brazil.
| | - Daniela Vieira Buchaim
- Medical School, Discipline of Human Morphophysiology, University of Marilia (UNIMAR), Av. Higino Muzi Filho, 1001 Campus Universitário, Jardim Araxa, Marília, São Paulo CEP 17525-902, Brazil.
- Medical School, Discipline of Neuroanatomy, University Center of Adamantina (UNIFAI), Rua Nove de Julho, 730, Centro, Adamantina, São Paulo CEP 17800-000, Brazil.
| | - Natália Kawano
- Medical School, Discipline of Human Morphophysiology, University of Marilia (UNIMAR), Av. Higino Muzi Filho, 1001 Campus Universitário, Jardim Araxa, Marília, São Paulo CEP 17525-902, Brazil.
| | - Gabriela Furlanette
- Medical School, Discipline of Human Morphophysiology, University of Marilia (UNIMAR), Av. Higino Muzi Filho, 1001 Campus Universitário, Jardim Araxa, Marília, São Paulo CEP 17525-902, Brazil.
| | - Karina Torres Pomini
- Department of Biological Sciences (Anatomy), Bauru School of Dentistry, University of São Paulo (USP), Alameda Dr. Octávio Pinheiro Brisola 9-75, Vila Nova Cidade Universitária, Bauru, São Paulo CEP 17012-901, Brazil.
| | - Rogério Leone Buchaim
- Department of Biological Sciences (Anatomy), Bauru School of Dentistry, University of São Paulo (USP), Alameda Dr. Octávio Pinheiro Brisola 9-75, Vila Nova Cidade Universitária, Bauru, São Paulo CEP 17012-901, Brazil.
- Medical School, Discipline of Human Morphophysiology, University of Marilia (UNIMAR), Av. Higino Muzi Filho, 1001 Campus Universitário, Jardim Araxa, Marília, São Paulo CEP 17525-902, Brazil.
| |
Collapse
|
36
|
Adamsky A, Goshen I. Astrocytes in Memory Function: Pioneering Findings and Future Directions. Neuroscience 2017; 370:14-26. [PMID: 28571720 DOI: 10.1016/j.neuroscience.2017.05.033] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/05/2017] [Accepted: 05/19/2017] [Indexed: 12/29/2022]
Abstract
Astrocytes have been generally believed to perform mainly homeostatic and supportive functions for neurons in the central nervous system. Recently, a growing body of evidence suggests previously unrecognized and surprising functions for astrocytes, including regulation of synaptic formation, transmission and plasticity, all of which are considered as the infrastructure for information processing and memory formation and stabilization. This review discusses the involvement of astrocytes in memory functions and the possible mechanisms that may underlie it. We review the important breakthroughs obtained in this field, as well as some of the controversies that arose from the past difficulty to manipulate these cells in a cell type-specific and non-invasive manner. Finally, we present new research avenues based on the advanced tools becoming available in recent years: optogenetics and chemogenetics, and the potential ways in which these tools may further illuminate the role of astrocytes in memory processes.
Collapse
Affiliation(s)
- Adar Adamsky
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Givat Ram, Jerusalem 91904, Israel
| | - Inbal Goshen
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Givat Ram, Jerusalem 91904, Israel.
| |
Collapse
|
37
|
Perez-Tilve D. Novel Hypothalamic Mechanisms in the Pathophysiological Control of Body Weight and Metabolism. Endocrinology 2017; 158:1085-1094. [PMID: 28200100 DOI: 10.1210/en.2016-1944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/03/2017] [Indexed: 12/31/2022]
Abstract
The incidence of obesity, with its impact on the development of comorbidities, including diabetes and cardiovascular disease, represents one of the greatest global health threats of the 21st century. This is particularly damning considering the vast progress that has been made in understanding the factors and molecular mechanisms playing a role in the control of energy balance by the central nervous system, especially during the past 3 decades. Despite the wealth of newfound knowledge, effective therapies for prevention of and/or intervention in obesity have not been forthcoming. That said, recent technological advances and the revisiting of previously discarded concepts have identified novel neural mechanisms involved in the control of energy homeostasis, thereby providing potential new targets and experimental approaches that may bring us closer to effective therapies to improve metabolic control. This review summarizes some of the most recent findings, with special emphasis on the role of neural circuits of the hypothalamus.
Collapse
Affiliation(s)
- Diego Perez-Tilve
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| |
Collapse
|