1
|
Eldar D, Albert S, Tatyana A, Galina S, Albert R, Yana M. Optogenetic approaches for neural tissue regeneration: A review of basic optogenetic principles and target cells for therapy. Neural Regen Res 2026; 21:521-533. [PMID: 39995064 DOI: 10.4103/nrr.nrr-d-24-00685] [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: 06/21/2024] [Accepted: 10/17/2024] [Indexed: 02/26/2025] Open
Abstract
Optogenetics has revolutionized the field of neuroscience by enabling precise control of neural activity through light-sensitive proteins known as opsins. This review article discusses the fundamental principles of optogenetics, including the activation of both excitatory and inhibitory opsins, as well as the development of optogenetic models that utilize recombinant viral vectors. A considerable portion of the article addresses the limitations of optogenetic tools and explores strategies to overcome these challenges. These strategies include the use of adeno-associated viruses, cell-specific promoters, modified opsins, and methodologies such as bioluminescent optogenetics. The application of viral recombinant vectors, particularly adeno-associated viruses, is emerging as a promising avenue for clinical use in delivering opsins to target cells. This trend indicates the potential for creating tools that offer greater flexibility and accuracy in opsin delivery. The adaptations of these viral vectors provide advantages in optogenetic studies by allowing for the restricted expression of opsins through cell-specific promoters and various viral serotypes. The article also examines different cellular targets for optogenetics, including neurons, astrocytes, microglia, and Schwann cells. Utilizing specific promoters for opsin expression in these cells is essential for achieving precise and efficient stimulation. Research has demonstrated that optogenetic stimulation of both neurons and glial cells-particularly the distinct phenotypes of microglia, astrocytes, and Schwann cells-can have therapeutic effects in neurological diseases. Glial cells are increasingly recognized as important targets for the treatment of these disorders. Furthermore, the article emphasizes the emerging field of bioluminescent optogenetics, which combines optogenetic principles with bioluminescent proteins to visualize and manipulate neural activity in real time. By integrating molecular genetics techniques with bioluminescence, researchers have developed methods to monitor neuronal activity efficiently and less invasively, enhancing our understanding of central nervous system function and the mechanisms of plasticity in neurological disorders beyond traditional neurobiological methods. Evidence has shown that optogenetic modulation can enhance motor axon regeneration, achieve complete sensory reinnervation, and accelerate the recovery of neuromuscular function. This approach also induces complex patterns of coordinated motor neuron activity and promotes neural reorganization. Optogenetic approaches hold immense potential for therapeutic interventions in the central nervous system. They enable precise control of neural circuits and may offer new treatments for neurological disorders, particularly spinal cord injuries, peripheral nerve injuries, and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Davletshin Eldar
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Sufianov Albert
- Department of Neurosurgery, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
- Research and Educational Institute of Neurosurgery, Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Ageeva Tatyana
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Sufianova Galina
- Department of Pharmacology, Tyumen State Medical University, Tyumen, Russia
| | - Rizvanov Albert
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Mukhamedshina Yana
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| |
Collapse
|
2
|
Furukawa K, Ikoma Y, Niino Y, Hiraoka Y, Tanaka K, Miyawaki A, Hirrlinger J, Matsui K. Dynamics of Neuronal and Astrocytic Energy Molecules in Epilepsy. J Neurochem 2025; 169:e70044. [PMID: 40108970 PMCID: PMC11923518 DOI: 10.1111/jnc.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
The dynamics of energy molecules in the mouse brain during metabolic challenges induced by epileptic seizures were examined. A transgenic mouse line expressing a fluorescence resonance energy transfer (FRET)-based adenosine triphosphate (ATP) sensor, selectively expressed in the cytosol of neurons, was used. An optical fiber was inserted into the hippocampus, and changes in cytosolic ATP concentration were estimated using the fiber photometry method. To induce epileptic neuronal hyperactivity, a train of electrical stimuli was delivered to a bipolar electrode placed alongside the optical fiber. Although maintaining a steady cytosolic ATP concentration is crucial for cell survival, a single episode of epileptic neuronal hyperactivity drastically reduced neuronal ATP levels. Interestingly, the magnitude of ATP reduction did not increase with the exacerbation of epilepsy, but rather decreased. This suggests that the primary consumption of ATP during epileptic neuronal hyperactivity may not be solely directed toward restoring the Na+ and K+ ionic imbalance caused by action potential bursts. Cytosolic ATP concentration reflects the balance between supply and consumption. To investigate the metabolic flux leading to neuronal ATP production, a new FRET-based pyruvate sensor was developed and selectively expressed in the cytosol of astrocytes in transgenic mice. Upon epileptic neuronal hyperactivity, an increase in astrocytic pyruvate concentration was observed. Changes in the supply of energy molecules, such as glucose and oxygen, due to blood vessel constriction or dilation, as well as metabolic alterations in astrocyte function, may contribute to cytosolic ATP dynamics in neurons.
Collapse
Affiliation(s)
- Kota Furukawa
- Super‐network Brain PhysiologyGraduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Yoko Ikoma
- Super‐network Brain PhysiologyGraduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Yusuke Niino
- Laboratory for Cell Function DynamicsRIKEN Center for Brain ScienceWako‐CityJapan
| | - Yuichi Hiraoka
- Laboratory of Molecular NeuroscienceMedical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo Institute of TechnologyTokyoJapan
- Laboratory of Genome Editing for Biomedical ResearchMedical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo Institute of TechnologyTokyoJapan
| | - Kohichi Tanaka
- Laboratory of Genome Editing for Biomedical ResearchMedical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo Institute of TechnologyTokyoJapan
| | - Atsushi Miyawaki
- Laboratory for Cell Function DynamicsRIKEN Center for Brain ScienceWako‐CityJapan
- Biotechnological Optics Research TeamRIKEN Center for Advanced PhotonicsWako‐CityJapan
| | - Johannes Hirrlinger
- Carl‐Ludwig‐Institute for Physiology, Faculty of MedicineLeipzig UniversityLeipzigGermany
- Department of NeurogeneticsMax‐Planck‐Institute for Multidisciplinary SciencesGöttingenGermany
| | - Ko Matsui
- Super‐network Brain PhysiologyGraduate School of Life Sciences, Tohoku UniversitySendaiJapan
- Super‐network Brain Physiology, Graduate School of MedicineTohoku UniversitySendaiJapan
| |
Collapse
|
3
|
Yamao H, Matsui K. Astrocytic determinant of the fate of long-term memory. Glia 2025; 73:309-329. [PMID: 39495149 DOI: 10.1002/glia.24636] [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: 06/10/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Abstract
While some vivid memories are unyielding and unforgettable, others fade with time. Astrocytes are recognized for their role in modulating the brain's environment and have recently been considered integral to the brain's information processing and memory formation. This suggests their potential roles in emotional perception and memory formation. In this study, we delve into the impact of amygdala astrocytes on fear behaviors and memory, employing astrocyte-specific optogenetic manipulations in mice. Our findings reveal that astrocytic photoactivation with channelrhodopsin-2 (ChR2) provokes aversive behavioral responses, while archaerhodopsin-T (ArchT) photoactivation diminishes fear perception. ChR2 photoactivation amplifies fear perception and fear memory encoding but obstructs its consolidation. On the other hand, ArchT photoactivation inhibits memory formation during intense aversive stimuli, possibly due to weakened fear perception. However, it prevents the decay of remote fear memory over three weeks. Crucially, these memory effects were observed when optogenetic manipulations coincided with the aversive experience, indicating a deterministic role of astrocytic states at the exact moment of fear experiences in shaping long-term memory. This research underscores the significant and multifaceted role of astrocytes in emotional perception, fear memory formation, and modulation, suggesting a sophisticated astrocyte-neuron communication mechanism underlying basic emotional state transitions of information processing in the brain.
Collapse
Affiliation(s)
- Hiroki Yamao
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
- Super-network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
4
|
Peña-Ortega F. Microglial modulation of neuronal network function and plasticity. J Neurophysiol 2025; 133:661-680. [PMID: 39819084 DOI: 10.1152/jn.00458.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/08/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS), which have been classically viewed as involved in CNS responses to damage and tissue repair. However, microglia are constantly sensing neuronal network activity and changes in the CNS milieu, establishing complex state-dependent microglia-neuron interactions that impact their functions. By doing so, microglia perform a wide range of physiological roles, including brain homeostasis maintenance, control of neural connectivity, network function modulation, as well as functional and morphological plasticity regulation in health and disease. Here, the author reviews recent evidence of the modulations induced by microglia, a highly heterogeneous cell type, on synaptic and intrinsic neuronal properties, and on neuronal network patterns during perinatal development and adulthood. The reviewed evidence clearly indicates that microglia are important, if not essential, for brain function and plasticity in both health and disease.
Collapse
Affiliation(s)
- Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Mexico
| |
Collapse
|
5
|
Suarez A, Fernandez L, Riera J. Characterizing astrocyte-mediated neurovascular coupling by combining optogenetics and biophysical modeling. J Cereb Blood Flow Metab 2025:271678X241311010. [PMID: 39791314 PMCID: PMC11719438 DOI: 10.1177/0271678x241311010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025]
Abstract
Vasoactive signaling from astrocytes is an important contributor to the neurovascular coupling (NVC), which aims at providing energy to neurons during brain activation by increasing blood perfusion in the surrounding vasculature. Pharmacological manipulations have been previously combined with experimental techniques (e.g., transgenic mice, uncaging, and multiphoton microscopy) and stimulation paradigms to isolate in vivo individual pathways of the astrocyte-mediated NVC. Unfortunately, these pathways are highly nonlinear and non-additive. To separate these pathways in a unified framework, we combine a comprehensive biophysical model of vasoactive signaling from astrocytes with a unique optogenetic stimulation method that selectively induces astrocytic Ca2+ signaling in a large population of astrocytes. We also use a sensitivity analysis and an optimization technique to estimate key model parameters. Optogenetically-induced Ca2+ signals in astrocytes cause a cerebral blood flow (CBF) response with two major components. Component-1 was rapid and smaller (ΔCBF∼13%, 18 seconds), while component-2 was slowest and highest (ΔCBF ∼18%, 45 seconds). The proposed biophysical model was adequate in reproducing component-2, which was validated with a pharmacological manipulation. Model's predictions were not in contradiction with previous studies. Finally, we discussed scenarios accounting for the existence of component-1, which once validated might be included in our model.
Collapse
Affiliation(s)
- Alejandro Suarez
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| | - Lazaro Fernandez
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| | - Jorge Riera
- Neuronal Mass Dynamics Lab, Department of Biomedical Engineering, Florida International, University, Miami, FL, USA
| |
Collapse
|
6
|
Liu S, Yang X, Zhao H, Zhao X, Fan K, Liu G, Li X, Du C, Liu J, Ma J. Cathepsin C exacerbates EAE by promoting the expansion of Tfh cells and the formation of TLSs in the CNS. Brain Behav Immun 2025; 123:123-142. [PMID: 39243987 DOI: 10.1016/j.bbi.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/05/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) mediated by CD4+ T helper (Th) cells, and characterized by immune cell infiltration, demyelination and neurodegeneration, with no definitive cure available. Thus, it is pivotal and imperative to acquire more profound comprehension of the underlying mechanisms implicated in MS. Dysregulated immune responses are widely believed to play a primary role in the pathogenesis of MS. Recently, a plethora of studies have demonstrated the involvement of T follicular helper (Tfh) cells and tertiary lymphoid-like structures (TLSs) in the pathogenesis and progression of MS. Cathepsin C (CatC) is a cysteine exopeptidase which is crucial for the activation of immune-cell-associated serine proteinases in many inflammatory diseases in peripheral system, such as rheumatoid arthritis and septicemia. We have previously demonstrated that CatC is involved in neuroinflammation and exacerbates demyelination in both cuprizone-induced and experimental autoimmune encephalomyelitis (EAE) mouse models. However, the underlying immunopathological mechanism remains elusive. In the present study, we established a recombinant myelin oligodendrocyte glycoprotein 35-55 peptide-induced EAE model using conditional CatC overexpression mice to investigate the effects of CatC on the alteration of CD4+ Th subsets, including Th1, Th2, Th17, Tfh and T regulatory cells. Our findings demonstrated that CatC particularly enhanced the population of Tfh cell in the brain, resulting in the earlier onset and more severe chronic syndrome of EAE. Furthermore, CatC promoted the formation of TLSs in the brain, leading to persistent neuroinflammation and exacerbating the severity of EAE in the chronic phase. Conversely, treatment with AZD7986, a specific inhibitor of CatC, effectively attenuated the syndrome of EAE and its effects caused by CatC both in vivo and in vitro. These findings provide a novel insight into the critical role of CatC in innate and adaptive immunity in EAE, and specific inhibitor of CatC, AZD7986, may contribute to potential therapeutic strategies for MS.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xiaohan Yang
- Department of Morphology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Henan Zhao
- Department of Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xinnan Zhao
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Kai Fan
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Gang Liu
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xia Li
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Cong Du
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Jianmei Ma
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning 116044, China.
| |
Collapse
|
7
|
Gobbo D, Kirchhoff F. Animal-based approaches to understanding neuroglia physiology in vitro and in vivo. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:229-263. [PMID: 40122627 DOI: 10.1016/b978-0-443-19104-6.00012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter describes the pivotal role of animal models for unraveling the physiology of neuroglial cells in the central nervous system (CNS). The two rodent species Mus musculus (mice) and Rattus norvegicus (rats) have been indispensable in scientific research due to their remarkable resemblance to humans anatomically, physiologically, and genetically. Their ease of maintenance, short gestation times, and rapid development make them ideal candidates for studying the physiology of astrocytes, oligodendrocyte-lineage cells, and microglia. Moreover, their genetic similarity to humans facilitates the investigation of molecular mechanisms governing neural physiology. Mice are largely the predominant model of neuroglial research, owing to advanced genetic manipulation techniques, whereas rats remain invaluable for applications requiring larger CNS structures for surgical manipulations. Next to rodents, other animal models, namely, Danio rerio (zebrafish) and Drosophila melanogaster (fruit fly), will be discussed to emphasize their critical role in advancing our understanding of glial physiology. Each animal model provides distinct advantages and disadvantages. By combining the strengths of each of them, researchers can gain comprehensive insights into glial function across species, ultimately promoting the understanding of glial physiology in the human CNS and driving the development of novel therapeutic interventions for CNS disorders.
Collapse
Affiliation(s)
- Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany.
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany; Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany.
| |
Collapse
|
8
|
Yokoyama K, Hiraoka Y, Abe Y, Tanaka KF. Visualization of myelin-forming oligodendrocytes in the adult mouse brain. J Neurochem 2025; 169:e16218. [PMID: 39233334 PMCID: PMC11657928 DOI: 10.1111/jnc.16218] [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: 03/26/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
Oligodendrocyte (OL) differentiation from oligodendrocyte precursor cells (OPCs) is considered to result in two populations: premyelinating and myelinating OLs. Recent single-cell RNA sequence data subdivided these populations into newly formed (NFOLs), myelin-forming (MFOLs), and mature (MOLs) oligodendrocytes. However, which newly proposed population corresponds to premyelinating or myelinating OLs is unknown. We focused on the NFOL-specific long non-coding oligodendrocyte 1 gene (LncOL1) and sought to label NFOLs under the control of the LncOL1 promoter using a tetracycline-controllable gene induction system. We demonstrated that LncOL1 was expressed by premyelinating OLs and that the MFOL-specific gene, Ctps, was not, indicating that NFOLs correspond to premyelinating OLs and that MFOLs and MOLs correspond to myelinating OLs. We then generated a LncOL1-tTA mouse in which a tetracycline transactivator (tTA) cassette was inserted downstream from the LncOL1 transcription initiation site. By crossing the LncOL1-tTA mice with tetO reporter mice, we generated LncOL1-tTA::tetO-yellow fluorescent protein (YFP) double-transgenic (LncOL1-YFP) mice. Although LncOL1 is non-coding, YFP was detected in LncOL1-YFP mice, indicating successful tTA translation. Unexpectedly, we found that the morphology of LncOL1-tTA-driven YFP+ cells was distinct from that of LncOL1+ premyelinating OLs and that the labeled cells instead appeared as myelinating OLs. We demonstrated from their RNA expression that YFP-labeled OLs were MFOLs, but not MOLs. Using the unique property of delayed YFP induction, we sought to determine whether MFOLs are constantly supplied from OPCs and differentiate into MOLs, or whether MFOLs pause their differentiation and sustain this stage in the adult brain. To achieve this objective, we irradiated adult LncOL1-YFP brains with X-rays to deplete dividing OPCs and their progeny. The irradiation extinguished YFP-labeled OLs, indicating that adult OPCs differentiated into MOLs during a single period. We established a new transgenic mouse line that genetically labels MFOLs, providing a reliable tool for investigating the dynamics of adult oligodendrogenesis.
Collapse
Affiliation(s)
- Kiichi Yokoyama
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| | - Yuichi Hiraoka
- Laboratory of Molecular NeuroscienceMedical Research Institute, Tokyo Medical and Dental UniversityTokyoJapan
- Laboratory of Genome Editing for Biomedical ResearchMedical Research Institute, Tokyo Medical and Dental UniversityTokyoJapan
| | - Yoshifumi Abe
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| | - Kenji F. Tanaka
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| |
Collapse
|
9
|
Vringer M, Zhou J, Gool JK, Bijlenga D, Lammers GJ, Fronczek R, Schinkelshoek MS. Recent insights into the pathophysiology of narcolepsy type 1. Sleep Med Rev 2024; 78:101993. [PMID: 39241492 DOI: 10.1016/j.smrv.2024.101993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/09/2024]
Abstract
Narcolepsy type 1 (NT1) is a sleep-wake disorder in which people typically experience excessive daytime sleepiness, cataplexy and other sleep-wake disturbances impairing daily life activities. NT1 symptoms are due to hypocretin deficiency. The cause for the observed hypocretin deficiency remains unclear, even though the most likely hypothesis is that this is due to an auto-immune process. The search for autoantibodies and autoreactive T-cells has not yet produced conclusive evidence for or against the auto-immune hypothesis. Other mechanisms, such as reduced corticotrophin-releasing hormone production in the paraventricular nucleus have recently been suggested. There is no reversive treatment, and the therapeutic approach is symptomatic. Early diagnosis and appropriate NT1 treatment is essential, especially in children to prevent impaired cognitive, emotional and social development. Hypocretin receptor agonists have been designed to replace the attenuated hypocretin signalling. Pre-clinical and clinical trials have shown encouraging initial results. A better understanding of NT1 pathophysiology may contribute to faster diagnosis or treatments, which may cure or prevent it.
Collapse
Affiliation(s)
- Marieke Vringer
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jingru Zhou
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jari K Gool
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands; Department of Anatomy & Neurosciences, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Compulsivity, Impulsivity and Attention, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Denise Bijlenga
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Gert Jan Lammers
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Rolf Fronczek
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Mink S Schinkelshoek
- Stichting Epilepsie Instellingen Nederland (SEIN), Sleep-Wake center, Heemstede, the Netherlands; Department of Neurology, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
10
|
Dantsuji M, Mochizuki A, Nakayama K, Kanamaru M, Izumizaki M, Tanaka KF, Inoue T, Nakamura S. Optogenetic activation of serotonergic neurons changes masticatory movement in freely moving mice. Sci Rep 2024; 14:27703. [PMID: 39533095 PMCID: PMC11557829 DOI: 10.1038/s41598-024-79429-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
The serotonergic system modulates the neural circuits involved in jaw movement; however, the role of serotonin (5-HT) neurons in masticatory movement remains unclear. Here, we investigated the effect of selective activation of 5-HT neurons in the dorsal raphe nucleus (DRN), or the raphe obscurus nucleus (ROb), on voluntary masticatory movement using transgenic mice expressing the channelrhodopsin-2 (ChR2) mutant (C128S) in central 5-HT neurons. During voluntary mastication, DRN blue light illumination increased masticatory frequency and decreased the root mean square peak amplitude of electromyography (EMG) in the masseter muscles. DRN blue light illumination also decreased EMG burst duration in the masseter and digastric muscles. These changes were blocked by a 5-HT2A receptor antagonist. Conversely, ROb blue light illumination during voluntary mastication did not affect masticatory frequency and EMG bursts in the masseter and digastric muscles. DRN or ROb blue light illumination during the resting state did not induce rhythmic jaw movement such as mastication but induced an increase in EMG activity in masseter and digastric muscles. These results suggest that both DRN and ROb 5-HT neurons may facilitate jaw movement. Furthermore, DRN 5-HT neuron may contribute to changes in masticatory patterns during the masticatory sequence.
Collapse
Affiliation(s)
- Masanori Dantsuji
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Ayako Mochizuki
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Kiyomi Nakayama
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Mitsuko Kanamaru
- Faculty of Arts and Sciences at Fujiyoshida, Showa University, Yamanashi, 403-0005, Japan
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Tomio Inoue
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai Shinagawa-ku, Tokyo, 142-8555, Japan
- Department of Contemporary life design, Kyoto Koka Women's University, Kyoto, 615-0882, Japan
| | - Shiro Nakamura
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai Shinagawa-ku, Tokyo, 142-8555, Japan.
| |
Collapse
|
11
|
Suzuki H, Murata J, Unekawa M, Kanno I, Izawa Y, Tomita Y, Tanaka KF, Nakahara J, Masamoto K. Microfluctuations in Capillary Lumens Independent of Pericyte Lining Density in the Anesthetized Mouse Cortex. Microcirculation 2024; 31:e12885. [PMID: 39283679 DOI: 10.1111/micc.12885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/25/2024] [Accepted: 08/28/2024] [Indexed: 11/12/2024]
Abstract
OBJECTIVE This study aimed to examine the spatiotemporal coherence of capillary lumen fluctuations in relation to spatial variations in the pericyte lining in the cortex of anesthetized mice. METHODS Two-photon microscopic angiography data (previously published) were reanalyzed, and spatial variations in capillary diameter fluctuations at rest and in capillary lining with vascular mural cells were measured along capillary centerlines. RESULTS Relatively large diameters of the capillaries (5.5 μm) coincided with a dense pericyte lining, while small capillaries (4.3 μm) had a sparse pericyte lining. Temporal variations had a frequency of about 0.1 Hz with an amplitude of 0.5 μm, which were negatively correlated with pericyte lining density. Spatial frequency analysis further revealed a common pattern of spatial variations in capillary diameter and pericyte lining, but temporal variations differed. The temporal variations in capillary lumens were locally distinct from those in neighboring locations, suggesting intrinsic fluctuations independent of the pericyte lining. CONCLUSIONS Capillary lumens in the brain exhibit slow microfluctuations that are independent of pericyte lining. These microfluctuations could affect the distribution of flowing blood cells and may be important for homogenizing their distribution in capillary networks.
Collapse
Affiliation(s)
- Hiroki Suzuki
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
| | - Juri Murata
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
| | - Miyuki Unekawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Iwao Kanno
- Department of Functional Brain Imaging Research, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yoshikane Izawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Tomita
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
- Tomita Hospital, Nagoya, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuto Masamoto
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
- Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Tokyo, Japan
| |
Collapse
|
12
|
Taira M, Miyazaki KW, Miyazaki K, Chen J, Okitsu-Sakurayama S, Chaudhary A, Nishio M, Miyake T, Yamanaka A, Tanaka KF, Doya K. The differential effect of optogenetic serotonergic manipulation on sustained motor actions and waiting for future rewards in mice. Front Neurosci 2024; 18:1433061. [PMID: 39385850 PMCID: PMC11461476 DOI: 10.3389/fnins.2024.1433061] [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: 05/15/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024] Open
Abstract
Serotonin is an essential neuromodulator that affects behavioral and cognitive functions. Previous studies have shown that activation of serotonergic neurons in the dorsal raphe nucleus (DRN) promotes patience to wait for future rewards. However, it is still unclear whether serotonergic neurons also regulate persistence to act for future rewards. Here we used optogenetic activation and inhibition of DRN serotonergic neurons to examine their effects on sustained motor actions for future rewards. We trained mice to perform waiting and repeated lever-pressing tasks with variable reward delays and tested effects of optogenetic activation and inhibition of DRN serotonergic neurons on task performance. Interestingly, in the lever-pressing task, mice tolerated longer delays as they repeatedly pressed a lever than in the waiting task, suggesting that lever-pressing actions may not simply be costly, but may also be subjectively rewarding. Optogenetic activation of DRN serotonergic neurons prolonged waiting duration in the waiting task, consistent with previous studies. However, its effect on lever presses was nuanced, and was detected only by focusing on the period before premature reward check and by subtracting the trends within and across sessions using generalized linear model. While optogenetic inhibition decreased waiting, it did not affect lever pressing time or numbers. These results revealed that the necessity of motor actions may increase motivation for delayed rewards and that DRN serotonergic neurons more significantly promote waiting rather than persistent motor actions for future rewards.
Collapse
Affiliation(s)
- Masakazu Taira
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Department of Psychology, University of Sydney, Camperdown, NSW, Australia
| | - Kayoko W. Miyazaki
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Katsuhiko Miyazaki
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Jianning Chen
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Shiho Okitsu-Sakurayama
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Anupama Chaudhary
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Mika Nishio
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- School of Medicine, Tohoku University, Sendai, Japan
| | - Tsukasa Miyake
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- School of Medicine, University of the Ryukyus, Okinawa, Japan
| | | | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Doya
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
13
|
Natsubori A, Kwon S, Honda Y, Kojima T, Karashima A, Masamoto K, Honda M. Serotonergic regulation of cortical neurovascular coupling and hemodynamics upon awakening from sleep in mice. J Cereb Blood Flow Metab 2024; 44:1591-1607. [PMID: 38477254 PMCID: PMC11418750 DOI: 10.1177/0271678x241238843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024]
Abstract
Neurovascular coupling (NVC) is the functional hyperemia of the brain responding to local neuronal activity. It is mediated by astrocytes and affected by subcortical ascending pathways in the cortex that convey information, such as sensory stimuli and the animal condition. Here, we investigate the influence of the raphe serotonergic system, a subcortical ascending arousal system in animals, on the modulation of cortical NVC and cerebral blood flow (CBF). Raphe serotonergic neurons were optogenically activated for 30 s, which immediately awakened the mice from non-rapid eye movement sleep. This caused a biphasic cortical hemodynamic change: a transient increase for a few seconds immediately after photostimulation onset, followed by a large progressive decrease during the stimulation period. Serotonergic neuron activation increased intracellular Ca2+ levels in cortical pyramidal neurons and astrocytes, demonstrating its effect on the NVC components. Pharmacological inhibition of cortical neuronal firing activity and astrocyte metabolic activity had small hypovolemic effects on serotonin-induced biphasic CBF changes, while blocking 5-HT1B receptors expressed primarily in cerebral vasculature attenuated the decreasing CBF phase. This suggests that serotonergic neuron activation leading to animal awakening could allow the NVC to exert a hyperemic function during a biphasic CBF response, with a predominant decrease in the cortex.
Collapse
Affiliation(s)
- Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Soojin Kwon
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yoshiko Honda
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takashi Kojima
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Akihiro Karashima
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai, Japan
| | - Kazuto Masamoto
- Dept. Mechanical and Intelligent Systems Engineering, Univ. of Electro-Communications, Tokyo, Japan
| | - Makoto Honda
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
14
|
Shigetomi E, Suzuki H, Hirayama YJ, Sano F, Nagai Y, Yoshihara K, Koga K, Tateoka T, Yoshioka H, Shinozaki Y, Kinouchi H, Tanaka KF, Bito H, Tsuda M, Koizumi S. Disease-relevant upregulation of P2Y 1 receptor in astrocytes enhances neuronal excitability via IGFBP2. Nat Commun 2024; 15:6525. [PMID: 39117630 PMCID: PMC11310333 DOI: 10.1038/s41467-024-50190-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 06/26/2024] [Indexed: 08/10/2024] Open
Abstract
Reactive astrocytes play a pivotal role in the pathogenesis of neurological diseases; however, their functional phenotype and the downstream molecules by which they modify disease pathogenesis remain unclear. Here, we genetically increase P2Y1 receptor (P2Y1R) expression, which is upregulated in reactive astrocytes in several neurological diseases, in astrocytes of male mice to explore its function and the downstream molecule. This astrocyte-specific P2Y1R overexpression causes neuronal hyperexcitability by increasing both astrocytic and neuronal Ca2+ signals. We identify insulin-like growth factor-binding protein 2 (IGFBP2) as a downstream molecule of P2Y1R in astrocytes; IGFBP2 acts as an excitatory signal to cause neuronal excitation. In neurological disease models of epilepsy and stroke, reactive astrocytes upregulate P2Y1R and increase IGFBP2. The present findings identify a mechanism underlying astrocyte-driven neuronal hyperexcitability, which is likely to be shared by several neurological disorders, providing insights that might be relevant for intervention in diverse neurological disorders.
Collapse
Affiliation(s)
- Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan.
| | - Hideaki Suzuki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Yukiho J Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Fumikazu Sano
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Yuki Nagai
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Kohei Yoshihara
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keisuke Koga
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Neurophysiology, Hyogo College of Medicine, Hyogo, 663-8501, Japan
| | - Toru Tateoka
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hideyuki Yoshioka
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hiroyuki Kinouchi
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan.
| |
Collapse
|
15
|
Araki S, Onishi I, Ikoma Y, Matsui K. Astrocyte switch to the hyperactive mode. Glia 2024; 72:1418-1434. [PMID: 38591259 DOI: 10.1002/glia.24537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/10/2024]
Abstract
Increasing pieces of evidence have suggested that astrocyte function has a strong influence on neuronal activity and plasticity, both in physiological and pathophysiological situations. In epilepsy, astrocytes have been shown to respond to epileptic neuronal seizures; however, whether they can act as a trigger for seizures has not been determined. Here, using the copper implantation method, spontaneous neuronal hyperactivity episodes were reliably induced during the week following implantation. With near 24-h continuous recording for over 1 week of the local field potential with in vivo electrophysiology and astrocyte cytosolic Ca2+ with the fiber photometry method, spontaneous occurrences of seizure episodes were captured. Approximately 1 day after the implantation, isolated aberrant astrocyte Ca2+ events were often observed before they were accompanied by neuronal hyperactivity, suggesting the role of astrocytes in epileptogenesis. Within a single developed episode, astrocyte Ca2+ increase preceded the neuronal hyperactivity by ~20 s, suggesting that actions originating from astrocytes could be the trigger for the occurrence of epileptic seizures. Astrocyte-specific stimulation by channelrhodopsin-2 or deep-brain direct current stimulation was capable of inducing neuronal hyperactivity. Injection of an astrocyte-specific metabolic inhibitor, fluorocitrate, was able to significantly reduce the magnitude of spontaneously occurring neuronal hyperactivity. These results suggest that astrocytes have a role in triggering individual seizures and the reciprocal astrocyte-neuron interactions likely amplify and exacerbate seizures. Therefore, future epilepsy treatment could be targeted at astrocytes to achieve epilepsy control.
Collapse
Affiliation(s)
- Shun Araki
- Super-network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Ichinosuke Onishi
- Super-network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yoko Ikoma
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
16
|
Tan W, Ikoma Y, Takahashi Y, Konno A, Hirai H, Hirase H, Matsui K. Anxiety control by astrocytes in the lateral habenula. Neurosci Res 2024; 205:1-15. [PMID: 38311032 DOI: 10.1016/j.neures.2024.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/06/2024]
Abstract
The potential role of astrocytes in lateral habenula (LHb) in modulating anxiety was explored in this study. The habenula are a pair of small nuclei located above the thalamus, known for their involvement in punishment avoidance and anxiety. Herein, we observed an increase in theta-band oscillations of local field potentials (LFPs) in the LHb when mice were exposed to anxiety-inducing environments. Electrical stimulation of LHb at theta-band frequency promoted anxiety-like behavior. Calcium (Ca2+) levels and pH in the cytosol of astrocytes and local brain blood volume changes were studied in mice expressing either a Ca2+ or a pH sensor protein specifically in astrocytes and mScarlet fluorescent protein in the blood plasma using fiber photometry. An acidification response to anxiety was observed. Photoactivation of archaerhopsin-T (ArchT), an optogenetic tool that acts as an outward proton pump, results in intracellular alkalinization. Photostimulation of LHb in astrocyte-specific ArchT-expressing mice resulted in dissipation of theta-band LFP oscillation in an anxiogenic environment and suppression of anxiety-like behavior. These findings provide evidence that LHb astrocytes modulate anxiety and may offer a new target for treatment of anxiety disorders.
Collapse
Affiliation(s)
- Wanqin Tan
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Yoko Ikoma
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Yusuke Takahashi
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan; Systems Bioinformatics, Graduate School of Information Sciences, Tohoku University, Sendai 980-8579 Japan
| | - Ayumu Konno
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi 371-8511, Gunma, Japan; Viral Vector Core, Gunma University Initiative for Advanced Research, Maebashi 371-8511, Gunma, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi 371-8511, Gunma, Japan; Viral Vector Core, Gunma University Initiative for Advanced Research, Maebashi 371-8511, Gunma, Japan
| | - Hajime Hirase
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan.
| |
Collapse
|
17
|
Hamada HT, Abe Y, Takata N, Taira M, Tanaka KF, Doya K. Optogenetic activation of dorsal raphe serotonin neurons induces brain-wide activation. Nat Commun 2024; 15:4152. [PMID: 38755120 PMCID: PMC11099070 DOI: 10.1038/s41467-024-48489-6] [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: 08/12/2022] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Serotonin is a neuromodulator that affects multiple behavioral and cognitive functions. Nonetheless, how serotonin causes such a variety of effects via brain-wide projections and various receptors remains unclear. Here we measured brain-wide responses to optogenetic stimulation of serotonin neurons in the dorsal raphe nucleus (DRN) of the male mouse brain using functional MRI with an 11.7 T scanner and a cryoprobe. Transient activation of DRN serotonin neurons caused brain-wide activation, including the medial prefrontal cortex, the striatum, and the ventral tegmental area. The same stimulation under anesthesia with isoflurane decreased brain-wide activation, including the hippocampal complex. These brain-wide response patterns can be explained by DRN serotonergic projection topography and serotonin receptor expression profiles, with enhanced weights on 5-HT1 receptors. Together, these results provide insight into the DR serotonergic system, which is consistent with recent discoveries of its functions in adaptive behaviors.
Collapse
Affiliation(s)
- Hiro Taiyo Hamada
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
- Research & Development Department, Araya Inc, Tokyo, Japan.
| | - Yoshifumi Abe
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Norio Takata
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Masakazu Taira
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Doya
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| |
Collapse
|
18
|
Komori T, Okamura K, Ikehara M, Yamamuro K, Endo N, Okumura K, Yamauchi T, Ikawa D, Ouji-Sageshima N, Toritsuka M, Takada R, Kayashima Y, Ishida R, Mori Y, Kamikawa K, Noriyama Y, Nishi Y, Ito T, Saito Y, Nishi M, Kishimoto T, Tanaka KF, Hiroi N, Makinodan M. Brain-derived neurotrophic factor from microglia regulates neuronal development in the medial prefrontal cortex and its associated social behavior. Mol Psychiatry 2024; 29:1338-1349. [PMID: 38243072 PMCID: PMC11189755 DOI: 10.1038/s41380-024-02413-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/21/2024]
Abstract
Microglia and brain-derived neurotrophic factor (BDNF) are essential for the neuroplasticity that characterizes critical developmental periods. The experience-dependent development of social behaviors-associated with the medial prefrontal cortex (mPFC)-has a critical period during the juvenile period in mice. However, whether microglia and BDNF affect social development remains unclear. Herein, we aimed to elucidate the effects of microglia-derived BDNF on social behaviors and mPFC development. Mice that underwent social isolation during p21-p35 had increased Bdnf in the microglia accompanied by reduced adulthood sociability. Additionally, transgenic mice overexpressing microglial Bdnf-regulated using doxycycline at different time points-underwent behavioral, electrophysiological, and gene expression analyses. In these mice, long-term overexpression of microglial BDNF impaired sociability and excessive mPFC inhibitory neuronal circuit activity. However, administering doxycycline to normalize BDNF from p21 normalized sociability and electrophysiological function in the mPFC, whereas normalizing BDNF from later ages (p45-p50) did not normalize electrophysiological abnormalities in the mPFC, despite the improved sociability. To evaluate the possible role of BDNF in human sociability, we analyzed the relationship between adverse childhood experiences and BDNF expression in human macrophages, a possible proxy for microglia. Results show that adverse childhood experiences positively correlated with BDNF expression in M2 but not M1 macrophages. In summary, our study demonstrated the influence of microglial BDNF on the development of experience-dependent social behaviors in mice, emphasizing its specific impact on the maturation of mPFC function, particularly during the juvenile period. Furthermore, our results propose a translational implication by suggesting a potential link between BDNF secretion from macrophages and childhood experiences in humans.
Collapse
Affiliation(s)
- Takashi Komori
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kazuya Okamura
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Minobu Ikehara
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Nozomi Endo
- Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kazuki Okumura
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Takahira Yamauchi
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Daisuke Ikawa
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | | | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Ryohei Takada
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yoshinori Kayashima
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Rio Ishida
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Mori
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kohei Kamikawa
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Noriyama
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yuki Nishi
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Yasuhiko Saito
- Department of Neurophysiology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Mayumi Nishi
- Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Noboru Hiroi
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX, 78229, USA
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, 78229, USA
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, 78229, USA
| | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University, Kashihara, Nara, 634-8521, Japan.
| |
Collapse
|
19
|
Asano Y, Sasaki D, Ikoma Y, Matsui K. Glial tone of aggression. Neurosci Res 2024; 202:39-51. [PMID: 38007191 DOI: 10.1016/j.neures.2023.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
Anger transition is often abrupt. In this study, we investigated the mechanisms responsible for switching and modulating aggression levels. The cerebellum is considered a center for motor coordination and learning; however, its connection to social behavior has long been observed. Here, we used the resident-intruder paradigm in male mice and examined local field potential (LFP) changes, glial cytosolic ion fluctuations, and vascular dynamics in the cerebellar vermis throughout various phases of a combat sequence. Notably, we observed the emergence of theta band oscillations in the LFP and sustained elevations in glial Ca2+ levels during combat breakups. When astrocytes, including Bergmann glial cells, were photoactivated using channelrhodopsin-2, the theta band emerged and an early combat breakup occurred. Within a single combat sequence, rapid alteration of offensive (fight) and passive (flight) responses were observed, which roughly correlated with decreases and increases in glial Ca2+, respectively. Neuron-glial interactions in the cerebellar vermis may play a role in adjusting Purkinje cell excitability and setting the tone of aggression. Future anger management strategies and clinical control of excessive aggression and violent behavior may be realized by developing a therapeutic strategy that adjusts glial activity in the cerebellum.
Collapse
Affiliation(s)
- Yuki Asano
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Daichi Sasaki
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Yoko Ikoma
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577 Japan.
| |
Collapse
|
20
|
Sasaki D, Imai K, Ikoma Y, Matsui K. Plastic vasomotion entrainment. eLife 2024; 13:RP93721. [PMID: 38629828 PMCID: PMC11023696 DOI: 10.7554/elife.93721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
Abstract
The presence of global synchronization of vasomotion induced by oscillating visual stimuli was identified in the mouse brain. Endogenous autofluorescence was used and the vessel 'shadow' was quantified to evaluate the magnitude of the frequency-locked vasomotion. This method allows vasomotion to be easily quantified in non-transgenic wild-type mice using either the wide-field macro-zoom microscopy or the deep-brain fiber photometry methods. Vertical stripes horizontally oscillating at a low temporal frequency (0.25 Hz) were presented to the awake mouse, and oscillatory vasomotion locked to the temporal frequency of the visual stimulation was induced not only in the primary visual cortex but across a wide surface area of the cortex and the cerebellum. The visually induced vasomotion adapted to a wide range of stimulation parameters. Repeated trials of the visual stimulus presentations resulted in the plastic entrainment of vasomotion. Horizontally oscillating visual stimulus is known to induce horizontal optokinetic response (HOKR). The amplitude of the eye movement is known to increase with repeated training sessions, and the flocculus region of the cerebellum is known to be essential for this learning to occur. Here, we show a strong correlation between the average HOKR performance gain and the vasomotion entrainment magnitude in the cerebellar flocculus. Therefore, the plasticity of vasomotion and neuronal circuits appeared to occur in parallel. Efficient energy delivery by the entrained vasomotion may contribute to meeting the energy demand for increased coordinated neuronal activity and the subsequent neuronal circuit reorganization.
Collapse
Affiliation(s)
- Daichi Sasaki
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Ken Imai
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Yoko Ikoma
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
| |
Collapse
|
21
|
Zhao S, Umpierre AD, Wu LJ. Tuning neural circuits and behaviors by microglia in the adult brain. Trends Neurosci 2024; 47:181-194. [PMID: 38245380 PMCID: PMC10939815 DOI: 10.1016/j.tins.2023.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/04/2023] [Accepted: 12/21/2023] [Indexed: 01/22/2024]
Abstract
Microglia are the primary immune cells of the CNS, contributing to both inflammatory damage and tissue repair in neurological disorder. In addition, emerging evidence highlights the role of homeostatic microglia in regulating neuronal activity, interacting with synapses, tuning neural circuits, and modulating behaviors. Herein, we review how microglia sense and regulate neuronal activity through synaptic interactions, thereby directly engaging with neural networks and behaviors. We discuss current studies utilizing microglial optogenetic and chemogenetic approaches to modulate adult neural circuits. These manipulations of microglia across different CNS regions lead to diverse behavioral consequences. We propose that spatial heterogeneity of microglia-neuron interaction lays the groundwork for understanding diverse functions of microglia in neural circuits and behaviors.
Collapse
Affiliation(s)
- Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | | | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Immunology, Mayo Clinic, Rochester, MN, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
22
|
Shinozaki Y, Namekata K, Guo X, Harada T. Glial cells as a promising therapeutic target of glaucoma: beyond the IOP. FRONTIERS IN OPHTHALMOLOGY 2024; 3:1310226. [PMID: 38983026 PMCID: PMC11182302 DOI: 10.3389/fopht.2023.1310226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/18/2023] [Indexed: 07/11/2024]
Abstract
Glial cells, a type of non-neuronal cell found in the central nervous system (CNS), play a critical role in maintaining homeostasis and regulating CNS functions. Recent advancements in technology have paved the way for new therapeutic strategies in the fight against glaucoma. While intraocular pressure (IOP) is the most well-known modifiable risk factor, a significant number of glaucoma patients have normal IOP levels. Because glaucoma is a complex, multifactorial disease influenced by various factors that contribute to its onset and progression, it is imperative that we consider factors beyond IOP to effectively prevent or slow down the disease's advancement. In the realm of CNS neurodegenerative diseases, glial cells have emerged as key players due to their pivotal roles in initiating and hastening disease progression. The inhibition of dysregulated glial function holds the potential to protect neurons and restore brain function. Consequently, glial cells represent an enticing therapeutic candidate for glaucoma, even though the majority of glaucoma research has historically concentrated solely on retinal ganglion cells (RGCs). In addition to the neuroprotection of RGCs, the proper regulation of glial cell function can also facilitate structural and functional recovery in the retina. In this review, we offer an overview of recent advancements in understanding the non-cell-autonomous mechanisms underlying the pathogenesis of glaucoma. Furthermore, state-of-the-art technologies have opened up possibilities for regenerating the optic nerve, which was previously believed to be incapable of regeneration. We will also delve into the potential roles of glial cells in the regeneration of the optic nerve and the restoration of visual function.
Collapse
Affiliation(s)
- Youichi Shinozaki
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
23
|
Iba C, Abe Y, Tanaka KF. Optogenetic demonstration of the involvement of SMA-negative mural cells in the regulation of cerebral blood flow. Front Physiol 2023; 14:1322250. [PMID: 38187133 PMCID: PMC10771846 DOI: 10.3389/fphys.2023.1322250] [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: 10/16/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Mural cells are critical components of the cerebral vasculature. They are categorized into three primary subsets: arteriole smooth muscle cells (aSMCs), pericytes (PCs) and venule smooth muscle cells (vSMCs). It is well known that aSMCs can directly regulate cerebral blood flow (CBF) with their own contraction and dilation mechanisms. On the other hand, the direct involvement of PCs or vSMCs in CBF regulation is controversial. This ambiguity is largely due to the lack of specifically manipulable tools to isolate their function. To address this issue, we employed a set-subtraction approach by using a combination of tTA-mediated gene induction and Cre-mediated gene excision. We developed transgenic mice expressing optical actuators, channelrhodopsin-2 (ChR2) and photoactivated adenylyl cyclase (PAC) in smooth muscle actin (SMA)-negative mural cells that lack the machinery for SMA-mediated vasoregulation. Using these mouse models, we assessed CBF alterations in response to optical stimulation using laser Doppler techniques. Our results showed that optical stimulation induced notable CBF changes in both models. This study provides evidence for the potential regulatory role of PCs and vSMCs in cerebral hemodynamics and introduces powerful tools to specifically manipulate these cell types in vascular neurobiology.
Collapse
Affiliation(s)
| | | | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Mukai Y, Okubo TS, Lazarus M, Ono D, Tanaka KF, Yamanaka A. Prostaglandin E 2 Induces Long-Lasting Inhibition of Noradrenergic Neurons in the Locus Coeruleus and Moderates the Behavioral Response to Stressors. J Neurosci 2023; 43:7982-7999. [PMID: 37734949 PMCID: PMC10669809 DOI: 10.1523/jneurosci.0353-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023] Open
Abstract
Neuronal activity is modulated not only by inputs from other neurons but also by various factors, such as bioactive substances. Noradrenergic (NA) neurons in the locus coeruleus (LC-NA neurons) are involved in diverse physiological functions, including sleep/wakefulness and stress responses. Previous studies have identified various substances and receptors that modulate LC-NA neuronal activity through techniques including electrophysiology, calcium imaging, and single-cell RNA sequencing. However, many substances with unknown physiological significance have been overlooked. Here, we established an efficient screening method for identifying substances that modulate LC-NA neuronal activity through intracellular calcium ([Ca2+]i) imaging using brain slices. Using both sexes of mice, we screened 53 bioactive substances, and identified five novel substances: gastrin-releasing peptide, neuromedin U, and angiotensin II, which increase [Ca2+]i, and pancreatic polypeptide and prostaglandin D2, which decrease [Ca2+]i Among them, neuromedin U induced the greatest response in female mice. In terms of the duration of [Ca2+]i change, we focused on prostaglandin E2 (PGE2), since it induces a long-lasting decrease in [Ca2+]i via the EP3 receptor. Conditional knock-out of the receptor in LC-NA neurons resulted in increased depression-like behavior, prolonged wakefulness in the dark period, and increased [Ca2+]i after stress exposure. Our results demonstrate the effectiveness of our screening method for identifying substances that modulate a specific neuronal population in an unbiased manner and suggest that stress-induced prostaglandin E2 can suppress LC-NA neuronal activity to moderate the behavioral response to stressors. Our screening method will contribute to uncovering previously unknown physiological functions of uncharacterized bioactive substances in specific neuronal populations.SIGNIFICANCE STATEMENT Bioactive substances modulate the activity of specific neuronal populations. However, since only a limited number of substances with predicted effects have been investigated, many substances that may modulate neuronal activity have gone unrecognized. Here, we established an unbiased method for identifying modulatory substances by measuring the intracellular calcium signal, which reflects neuronal activity. We examined noradrenergic (NA) neurons in the locus coeruleus (LC-NA neurons), which are involved in diverse physiological functions. We identified five novel substances that modulate LC-NA neuronal activity. We also found that stress-induced prostaglandin E2 (PGE2) may suppress LC-NA neuronal activity and influence behavioral outcomes. Our screening method will help uncover previously overlooked functions of bioactive substances and provide insight into unrecognized roles of specific neuronal populations.
Collapse
Affiliation(s)
- Yasutaka Mukai
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Tatsuo S Okubo
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS) and Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Chinese Institute for Brain Research, Beijing 102206, China
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| |
Collapse
|
25
|
Yoshida K, Kato D, Sugio S, Takeda I, Wake H. Activity-dependent oligodendrocyte calcium dynamics and their changes in Alzheimer's disease. Front Cell Neurosci 2023; 17:1154196. [PMID: 38026691 PMCID: PMC10644703 DOI: 10.3389/fncel.2023.1154196] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Oligodendrocytes (OCs) form myelin around axons, which is dependent on neuronal activity. This activity-dependent myelination plays a crucial role in training and learning. Previous studies have suggested that neuronal activity regulates proliferation and differentiation of oligodendrocyte precursor cells (OPCs) and myelination. In addition, deficient activity-dependent myelination results in impaired motor learning. However, the functional response of OC responsible for neuronal activity and their pathological changes is not fully elucidated. In this research, we aimed to understand the activity-dependent OC responses and their different properties by observing OCs using in vivo two-photon microscopy. We clarified that the Ca2+ activity in OCs is neuronal activity dependent and differentially regulated by neurotransmitters such as glutamate or adenosine triphosphate (ATP). Furthermore, in 5-month-old mice models of Alzheimer's disease, a period before the appearance of behavioral abnormalities, the elevated Ca2+ responses in OCs are ATP dependent, suggesting that OCs receive ATP from damaged tissue. We anticipate that our research will help in determining the correct therapeutic strategy for neurodegenerative diseases beyond the synapse.
Collapse
Affiliation(s)
- Kenji Yoshida
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
| | - Shouta Sugio
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
| | - Ikuko Takeda
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, National Institute of Natural Sciences, Okazaki, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
26
|
Ito H, Fukatsu N, Rahaman SM, Mukai Y, Izawa S, Ono D, Kilduff TS, Yamanaka A. Deficiency of orexin signaling during sleep is involved in abnormal REM sleep architecture in narcolepsy. Proc Natl Acad Sci U S A 2023; 120:e2301951120. [PMID: 37796986 PMCID: PMC10576136 DOI: 10.1073/pnas.2301951120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/10/2023] [Indexed: 10/07/2023] Open
Abstract
Narcolepsy is a sleep disorder caused by deficiency of orexin signaling. However, the neural mechanisms by which deficient orexin signaling causes the abnormal rapid eye movement (REM) sleep characteristics of narcolepsy, such as cataplexy and frequent transitions to REM states, are not fully understood. Here, we determined the activity dynamics of orexin neurons during sleep that suppress the abnormal REM sleep architecture of narcolepsy. Orexin neurons were highly active during wakefulness, showed intermittent synchronous activity during non-REM (NREM) sleep, were quiescent prior to the transition from NREM to REM sleep, and a small subpopulation of these cells was active during REM sleep. Orexin neurons that lacked orexin peptides were less active during REM sleep and were mostly silent during cataplexy. Optogenetic inhibition of orexin neurons established that the activity dynamics of these cells during NREM sleep regulate NREM-REM sleep transitions. Inhibition of orexin neurons during REM sleep increased subsequent REM sleep in "orexin intact" mice and subsequent cataplexy in mice lacking orexin peptides, indicating that the activity of a subpopulation of orexin neurons during the preceding REM sleep suppresses subsequent REM sleep and cataplexy. Thus, these results identify how deficient orexin signaling during sleep results in the abnormal REM sleep architecture characteristic of narcolepsy.
Collapse
Affiliation(s)
- Hiroto Ito
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya466-8550, Japan
- Japan Society for the Promotion of Science Research Fellowship for Young Scientists, Tokyo102-0083, Japan
| | - Noriaki Fukatsu
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya466-8550, Japan
| | - Sheikh Mizanur Rahaman
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya466-8550, Japan
| | - Yasutaka Mukai
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya466-8550, Japan
| | - Shuntaro Izawa
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya466-8550, Japan
| | - Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya466-8550, Japan
| | - Thomas S. Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA94025
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya464-8601, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya466-8550, Japan
- Chinese Institute for Brain Research, Beijing102206, China
- National Institute for Physiological Sciences, Aichi444-8585, Japan
- National Institutes of Natural Sciences, Aichi444-8585, Japan
- Division of Brain Sciences Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo160-8582, Japan
| |
Collapse
|
27
|
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
|
28
|
Katada Y, Kunimi H, Serizawa N, Lee D, Kobayashi K, Negishi K, Okano H, Tanaka KF, Tsubota K, Kurihara T. Starburst amacrine cells amplify optogenetic visual restoration through gap junctions. Mol Ther Methods Clin Dev 2023; 30:1-13. [PMID: 37324975 PMCID: PMC10265492 DOI: 10.1016/j.omtm.2023.05.011] [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: 07/22/2022] [Accepted: 05/09/2023] [Indexed: 06/17/2023]
Abstract
Ectopic induction of optogenetic actuators, such as channelrhodopsin, is a promising approach to restoring vision in the degenerating retina. However, the cell type-specific response of ectopic photoreception has not been well understood. There are limits to obtaining efficient gene expression in a specifically targeted cell population by a transgenic approach. In the present study, we established a murine model with high efficiency of gene induction to retinal ganglion cells (RGCs) and amacrine cells using an improved tetracycline transactivator-operator bipartite system (KENGE-tet system). To investigate the cell type-specific visual restorative effect, we expressed the channelrhodopsin gene into RGCs and amacrine cells using the KENGE-tet system. As a result, enhancement in the visual restorative effect was observed to RGCs and starburst amacrine cells. In conclusion, a photoresponse from amacrine cells may enhance the maintained response of RGCs and further increase or improve the visual restorative effect.
Collapse
Affiliation(s)
- Yusaku Katada
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiromitsu Kunimi
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Naho Serizawa
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Nutritional Sciences, Toyo University, Kita-ku, Tokyo 115-8650, Japan
| | - Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuo Tsubota
- Tsubota Laboratory, Inc, Shinjuku-ku, Tokyo 160-0016, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
29
|
Kono A, Shikano Y, Tanaka KF, Yamaura K, Tsutsui‐Kimura I. Inhibition of the dorsomedial striatal direct pathway is essential for the execution of action sequences. Neuropsychopharmacol Rep 2023; 43:414-424. [PMID: 37553985 PMCID: PMC10496086 DOI: 10.1002/npr2.12369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 08/10/2023] Open
Abstract
Contrary to the previous notion that the dorsomedial striatum (DMS) is crucial for acquiring new learning, accumulated evidence has suggested that the DMS also plays a role in the execution of already learned action sequences. Here, we examined how the direct and indirect pathways in the DMS regulate action sequences using a task that requires animals to press a lever consecutively. Cell-type-specific bulk Ca2+ recording revealed that the direct pathway was inhibited at the time of sequence execution. The sequence-related response was blunted in trials where the sequential behaviors were disrupted. Optogenetic activation at the sequence start caused distraction of action sequences without affecting motor function or memory of the task structure. By contrast with the direct pathway, the indirect pathway was slightly activated at the start of the sequence, but the optogenetic suppression of such sequence-related signaling did not impact the behaviors. These results suggest that the inhibition of the DMS direct pathway promotes sequence execution potentially by suppressing the formation of a new association.
Collapse
Affiliation(s)
- Anna Kono
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
- Division of Social Pharmacy, Center for Social Pharmacy and Pharmaceutical Care SciencesKeio University Faculty of PharmacyTokyoJapan
| | - Yu Shikano
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| | - Kenji F. Tanaka
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| | - Katsunori Yamaura
- Division of Social Pharmacy, Center for Social Pharmacy and Pharmaceutical Care SciencesKeio University Faculty of PharmacyTokyoJapan
| | - Iku Tsutsui‐Kimura
- Division of Brain SciencesInstitute for Advanced Medical Research, Keio University School of MedicineTokyoJapan
| |
Collapse
|
30
|
Murata J, Unekawa M, Kudo Y, Kotani M, Kanno I, Izawa Y, Tomita Y, Tanaka KF, Nakahara J, Masamoto K. Acceleration of the Development of Microcirculation Embolism in the Brain due to Capillary Narrowing. Stroke 2023; 54:2135-2144. [PMID: 37309687 DOI: 10.1161/strokeaha.122.042416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 05/05/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND Cerebral microvascular obstruction is critically involved in recurrent stroke and decreased cerebral blood flow with age. The obstruction must occur in the capillary with a greater resistance to perfusion pressure through the microvascular networks. However, little is known about the relationship between capillary size and embolism formation. This study aimed to determine whether the capillary lumen space contributes to the development of microcirculation embolism. METHODS To spatiotemporally manipulate capillary diameters in vivo, transgenic mice expressing the light-gated cation channel protein ChR2 (channelrhodopsin-2) in mural cells were used. The spatiotemporal changes in the regional cerebral blood flow in response to the photoactivation of ChR2 mural cells were first characterized using laser speckle flowgraphy. Capillary responses to optimized photostimulation were then examined in vivo using 2-photon microscopy. Finally, microcirculation embolism due to intravenously injected fluorescent microbeads was compared under conditions with or without photoactivation of ChR2 mural cells. RESULTS Following transcranial photostimulation, the stimulation intensity-dependent decrease in cerebral blood flow centered at the irradiation was observed (14%-49% decreases relative to the baseline). The cerebrovascular response to photostimulation showed significant constriction of the cerebral arteries and capillaries but not of the veins. As a result of vasoconstriction, a temporal stall of red blood cell flow occurred in the capillaries of the venous sides. The 2-photon excitation of a single ChR2 pericyte demonstrated the partial shrinkage of capillaries (7% relative to the baseline) around the stimulated cell. With the intravenous injection of microbeads, the occurrence of microcirculation embolism was significantly enhanced (11% increases compared to the control) with photostimulation. CONCLUSIONS Capillary narrowing increases the risk of developing microcirculation embolism in the venous sides of the cerebral capillaries.
Collapse
Affiliation(s)
- Juri Murata
- Graduate School of Informatics and Engineering (J.M., Y.K., M.K., K.M.), University of Electro-Communications, Tokyo, Japan
| | - Miyuki Unekawa
- Department of Neurology (M.U., Y.I., Y.T., J.N.), Keio University School of Medicine, Tokyo, Japan
| | - Yuya Kudo
- Graduate School of Informatics and Engineering (J.M., Y.K., M.K., K.M.), University of Electro-Communications, Tokyo, Japan
| | - Maho Kotani
- Graduate School of Informatics and Engineering (J.M., Y.K., M.K., K.M.), University of Electro-Communications, Tokyo, Japan
| | - Iwao Kanno
- Department of Functional Brain Imaging, National Institutes for Quantum Science and Technology, Chiba, Japan (I.K.)
| | - Yoshikane Izawa
- Department of Neurology (M.U., Y.I., Y.T., J.N.), Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Tomita
- Department of Neurology (M.U., Y.I., Y.T., J.N.), Keio University School of Medicine, Tokyo, Japan
- Tomita Hospital, Aichi, Japan (Y.T.)
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research (K.F.T.), Keio University School of Medicine, Tokyo, Japan
| | - Jin Nakahara
- Department of Neurology (M.U., Y.I., Y.T., J.N.), Keio University School of Medicine, Tokyo, Japan
| | - Kazuto Masamoto
- Graduate School of Informatics and Engineering (J.M., Y.K., M.K., K.M.), University of Electro-Communications, Tokyo, Japan
- Center for Neuroscience and Biomedical Engineering (K.M.), University of Electro-Communications, Tokyo, Japan
| |
Collapse
|
31
|
Aimi T, Matsuda K, Yuzaki M. C1ql1-Bai3 signaling is necessary for climbing fiber synapse formation in mature Purkinje cells in coordination with neuronal activity. Mol Brain 2023; 16:61. [PMID: 37488606 PMCID: PMC10367388 DOI: 10.1186/s13041-023-01048-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023] Open
Abstract
Changes in neural activity induced by learning and novel environments have been reported to lead to the formation of new synapses in the adult brain. However, the underlying molecular mechanism is not well understood. Here, we show that Purkinje cells (PCs), which have established adult-type monosynaptic innervation by climbing fibers (CFs) after elimination of weak CFs during development, can be reinnervated by multiple CFs by increased expression of the synaptic organizer C1ql1 in CFs or Bai3, a receptor for C1ql1, in PCs. In the adult cerebellum, CFs are known to have transverse branches that run in a mediolateral direction without forming synapses with PCs. Electrophysiological, Ca2+-imaging and immunohistochemical studies showed that overexpression of C1ql1 or Bai3 caused these CF transverse branches to elongate and synapse on the distal dendrites of mature PCs. Mature PCs were also reinnervated by multiple CFs when the glutamate receptor GluD2, which is essential for the maintenance of synapses between granule cells and PCs, was deleted. Interestingly, the effect of GluD2 knockout was not observed in Bai3 knockout PCs. In addition, C1ql1 levels were significantly upregulated in CFs of GluD2 knockout mice, suggesting that endogenous, not overexpressed, C1ql1-Bai3 signaling could regulate the reinnervation of mature PCs by CFs. Furthermore, the effects of C1ql1 and Bai3 overexpression required neuronal activity in the PC and CF, respectively. C1ql1 immunoreactivity at CF-PC synapses was reduced when the neuronal activity of CFs was suppressed. These results suggest that C1ql1-Bai3 signaling may mediate CF synaptogenesis in mature PCs, potentially in concert with neuronal activity.
Collapse
Affiliation(s)
- Takahiro Aimi
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Keiko Matsuda
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| |
Collapse
|
32
|
Makinodan M, Komori T, Okamura K, Ikehara M, Yamamuro K, Endo N, Okumura K, Yamauchi T, Ikawa D, Ouji-Sageshima N, Toritsuka M, Takada R, Kayashima Y, Ishida R, Mori Y, Kamikawa K, Noriyama Y, Nishi Y, Ito T, Saito Y, Nishi M, Kishimoto T, Tanaka K, Hiroi N. Brain-derived neurotrophic factor from microglia regulates neuronal development in the medial prefrontal cortex and its associated social behavior. RESEARCH SQUARE 2023:rs.3.rs-3094335. [PMID: 37461488 PMCID: PMC10350236 DOI: 10.21203/rs.3.rs-3094335/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Microglia and brain-derived neurotrophic factor (BDNF) are essential for the neuroplasticity that characterizes critical developmental periods. The experience-dependent development of social behaviors-associated with the medial prefrontal cortex (mPFC)-has a critical period during the juvenile period in mice. However, whether microglia and BDNF affect social development remains unclear. Herein, we aimed to elucidate the effects of microglia-derived BDNF on social behaviors and mPFC development. Mice that underwent social isolation during p21-p35 had increased Bdnf in the microglia accompanied by reduced adulthood sociability. Additionally, transgenic mice overexpressing microglia Bdnf-regulated using doxycycline at different time points-underwent behavioral, electrophysiological, and gene expression analyses. In these mice, long-term overexpression of microglia BDNF impaired sociability and excessive mPFC inhibitory neuronal circuit activity. However, administration of doxycycline to normalize BDNF from p21 normalized sociability and electrophysiological functions; this was not observed when BDNF was normalized from a later age (p45-p50). To evaluate the possible role of BDNF in human sociability, we analyzed the relationship between adverse childhood experiences and BDNF expression in human macrophages, a possible substitute for microglia. Results show that adverse childhood experiences positively correlated with BDNF expression in M2 but not M1 macrophages. Thus, microglia BDNF might regulate sociability and mPFC maturation in mice during the juvenile period. Furthermore, childhood experiences in humans may be related to BDNF secretion from macrophages.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - T Ito
- Keio University School of Medicine
| | | | | | | | | | - Noboru Hiroi
- University of Texas Health Science Center at San Antonio
| |
Collapse
|
33
|
Kanaya T, Ito R, Morizawa YM, Sasaki D, Yamao H, Ishikane H, Hiraoka Y, Tanaka K, Matsui K. Glial modulation of the parallel memory formation. Glia 2023. [PMID: 37364894 DOI: 10.1002/glia.24431] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 06/04/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023]
Abstract
Actions from glial cells could affect the readiness and efficacy of learning and memory. Using a mouse cerebellar-dependent horizontal optokinetic response motor learning paradigm, short-term memory (STM) formation during the online training period and long-term memory (LTM) formation during the offline rest period were studied. A large variability of online and offline learning efficacies was found. The early bloomers with booming STM often had a suppressed LTM formation and late bloomers with no apparent acute training effect often exhibited boosted offline learning performance. Anion channels containing LRRC8A are known to release glutamate. Conditional knockout of LRRC8A specifically in astrocytes including cerebellar Bergmann glia resulted in a complete loss of STM formation while the LTM formation during the rest period remained. Optogenetic manipulation of glial activity by channelrhodopsin-2 or archaerhodopsin-T (ArchT) during the online training resulted in enhancement or suppression of STM formation, respectively. STM and LTM are likely to be triggered simultaneously during online training, but LTM is expressed later during the offline period. STM appears to be volatile and the achievement during the online training is not handed over to LTM. In addition, we found that glial ArchT photoactivation during the rest period resulted in the augmentation of LTM formation. These data suggest that STM formation and LTM formation are parallel separate processes. Strategies to weigh more on the STM or the LTM could depend on the actions of the glial cells.
Collapse
Affiliation(s)
- Teppei Kanaya
- Super-Network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Ryo Ito
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Yosuke M Morizawa
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Daichi Sasaki
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Hiroki Yamao
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Hiroshi Ishikane
- Department of Psychology, Graduate School of Humanities, Senshu University, Kawasaki, Japan
| | - Yuichi Hiraoka
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute (MRI), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ko Matsui
- Super-Network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai, Japan
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
34
|
Chen LQ, Lv XJ, Guo QH, Lv SS, Lv N, Xu WD, Yu J, Zhang YQ. Asymmetric activation of microglia in the hippocampus drives anxiodepressive consequences of trigeminal neuralgia in rodents. Br J Pharmacol 2023; 180:1090-1113. [PMID: 36443951 DOI: 10.1111/bph.15994] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/26/2022] [Accepted: 11/14/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Patients suffering from trigeminal neuralgia are often accompanied by anxiety and depression. Microglia-mediated neuroinflammation is involved in the development of neuropathic pain and anxiodepression pathogenesis. Whether and how microglia are involved in trigeminal neuralgia-induced anxiodepression remains unclear. EXPERIMENTAL APPROACH Unilateral constriction of the infraorbital nerve (CION) was performed to establish trigeminal neuralgia in rat and mouse models. Mechanical allodynia and anxiodepressive-like behaviours were measured. Optogenetic and pharmacological manipulations were employed to investigate the role of hippocampal microglia in anxiety and depression caused by trigeminal neuralgia. KEY RESULTS Trigeminal neuralgia activated ipsilateral but not contralateral hippocampal microglia, up-regulated ipsilateral hippocampal ATP and interleukin-1β (IL-1β) levels, impaired ipsilateral hippocampal long-term potentiation (LTP) and induced anxiodepressive-like behaviours in a time-dependent manner in rodents. Pharmacological or optogenetic inhibition of ipsilateral hippocampal microglia completely blocked trigeminal neuralgia-induced anxiodepressive-like behaviours. Activation of unilateral hippocampal microglia directly elicited an anxiodepressive state and impaired hippocampal LTP. Knockdown of ipsilateral hippocampal P2X7 receptors prevented trigeminal neuralgia-induced microglial activation and anxiodepressive-like behaviours. Furthermore, we demonstrated that microglia-derived IL-1β mediated microglial activation-induced anxiodepressive-like behaviours and LTP impairment. CONCLUSION AND IMPLICATIONS These findings suggest that priming of microglia with ATP/P2X7 receptors in the ipsilateral hippocampus drives pain-related anxiodepressive-like behaviours via IL-1β. An asymmetric role of the bilateral hippocampus in trigeminal neuralgia-induced anxiety and depression was uncovered. The approaches targeting microglia and P2X7 signalling might offer novel therapies for trigeminal neuralgia-related anxiety and depressive disorder.
Collapse
Affiliation(s)
- Li-Qiang Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xue-Jing Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qing-Huan Guo
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Su-Su Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ning Lv
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Wen-Dong Xu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China.,Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Ikoma Y, Sasaki D, Matsui K. Local brain environment changes associated with epileptogenesis. Brain 2023; 146:576-586. [PMID: 36423658 DOI: 10.1093/brain/awac355] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/30/2022] [Accepted: 09/08/2022] [Indexed: 11/27/2022] Open
Abstract
Plastic change of the neuronal system has traditionally been assumed to be governed primarily by the long-term potentiation/depression mechanisms of synaptic transmission. However, a rather simple shift in the ambient ion, transmitter and metabolite concentrations could have a pivotal role in generating plasticity upon the physiological process of learning and memory. Local brain environment and metabolic changes could also be the cause and consequences of the pathogenesis leading to epilepsy. Governing of the local brain environment is the primal function of astrocytes. The metabolic state of the entire brain is strongly linked to the activity of the lateral hypothalamus. In this study, plastic change of astrocyte reactions in the lateral hypothalamus was examined using epileptogenesis as an extreme form of plasticity. Fluorescent sensors for calcium or pH expressed in astrocytes were examined for up to one week by in vivo fibre photometry in freely moving transgenic male mice. Optical fluctuations on a timescale of seconds is difficult to assess because these signals are heavily influenced by local brain blood volume changes and pH changes. Using a newly devised method for the analysis of the optical signals, changes in Ca2+ and pH in astrocytes and changes in local brain blood volume associated with hippocampal-stimulated epileptic seizures were extracted. Following a transient alkaline shift in the astrocyte triggered by neuronal hyperactivity, a prominent acidic shift appeared in response to intensified seizure which developed with kindling. The acidic shift was unexpected as transient increase in local brain blood volume was observed in response to intensified seizures, which should lead to efficient extrusion of the acidic CO2. The acidic shift could be a result of glutamate transporter activity and/or due to the increased metabolic load of astrocytes leading to increased CO2 and lactate production. This acidic shift may trigger additional gliotransmitter release from astrocytes leading to the exacerbation of epilepsy. As all cellular enzymic reactions are influenced by Ca2+ and pH, changes in these parameters could also have an impact on the neuronal circuit activity. Thus, controlling the astrocyte pH and/or Ca2+ could be a new therapeutic target for treatment of epilepsy or prevention of undesired plasticity associated with epileptogenesis.
Collapse
Affiliation(s)
- Yoko Ikoma
- Super-network Brain Physiology Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Daichi Sasaki
- Super-network Brain Physiology Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Ko Matsui
- Super-network Brain Physiology Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan.,Super-network Brain Physiology, Graduate School of Medicine, Tohoku University, Sendai 980-8577, Japan
| |
Collapse
|
36
|
Shigetomi E, Koizumi S. The role of astrocytes in behaviors related to emotion and motivation. Neurosci Res 2023; 187:21-39. [PMID: 36181908 DOI: 10.1016/j.neures.2022.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 10/14/2022]
Abstract
Astrocytes are present throughout the brain and intimately interact with neurons and blood vessels. Three decades of research have shown that astrocytes reciprocally communicate with neurons and other non-neuronal cells in the brain and dynamically regulate cell function. Astrocytes express numerous receptors for neurotransmitters, neuromodulators, and cytokines and receive information from neurons, other astrocytes, and other non-neuronal cells. Among those receptors, the main focus has been G-protein coupled receptors. Activation of G-protein coupled receptors leads to dramatic changes in intracellular signaling (Ca2+ and cAMP), which is considered a form of astrocyte activity. Methodological improvements in measurement and manipulation of astrocytes have advanced our understanding of the role of astrocytes in circuits and have begun to reveal unexpected functions of astrocytes in behavior. Recent studies have suggested that astrocytic activity regulates behavior flexibility, such as coping strategies for stress exposure, and plays an important role in behaviors related to emotion and motivation. Preclinical evidence suggests that impairment of astrocytic function contributes to psychiatric diseases, especially major depression. Here, we review recent progress on the role of astrocytes in behaviors related to emotion and motivation.
Collapse
Affiliation(s)
- Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan; Yamanashi GLIA Center, Graduate School of Medical Science, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan.
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan; Yamanashi GLIA Center, Graduate School of Medical Science, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Japan.
| |
Collapse
|
37
|
Oishi M, Passlick S, Yamazaki Y, Unekawa M, Adachi R, Yamada M, Imayoshi I, Abe Y, Steinhäuser C, Tanaka KF. Separate optogenetic manipulation of Nerve/glial antigen 2 (NG2) glia and mural cells using the NG2 promoter. Glia 2023; 71:317-333. [PMID: 36165697 DOI: 10.1002/glia.24273] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022]
Abstract
Nerve/glial antigen 2 (NG2) is a protein marker of NG2 glia and mural cells, and NG2 promoter activity is utilized to target these cells. However, the NG2 promoter cannot target NG2 glia and mural cells separately. This has been an obstacle for NG2 glia-specific manipulation. Here, we developed transgenic mice in which either cell type can be targeted using the NG2 promoter. We selected a tetracycline-controllable gene induction system for cell type-specific transgene expression, and generated NG2-tetracycline transactivator (tTA) transgenic lines. We crossed tTA lines with the tetO-ChR2 (channelrhodopsin-2)-EYFP line to characterize tTA-dependent transgene induction. We isolated two unique NG2-tTA mouse lines: one that induced ChR2-EYFP only in mural cells, likely due to the chromosomal position effect of NG2-tTA insertion, and the other that induced it in both cell types. We then applied a Cre-mediated set-subtraction strategy to the latter case and eliminated ChR2-EYFP from mural cells, resulting in NG2 glia-specific transgene induction. We further demonstrated that tTA-dependent ChR2 expression could manipulate cell function. Optogenetic mural cell activation decreased cerebral blood flow, as previously reported, indicating that tTA-mediated ChR2 expression was sufficient to impact cellular function. ChR2-mediated depolarization was observed in NG2 glia in acute hippocampal slices. In addition, ChR2-mediated depolarization of NG2 glia inhibited their proliferation but promoted their differentiation in juvenile mice. Since the tTA-tetO combination is expandable, the mural cell-specific NG2-tTA line and the NG2 glia-specific NG2-tTA line will permit us to conduct observational and manipulation studies to examine in vivo function of these cells separately.
Collapse
Affiliation(s)
- Mitsuhiro Oishi
- Division of Brain Sciences, Keio University School of Medicine, Tokyo, Japan
| | - Stefan Passlick
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Yoshihiko Yamazaki
- Department of Physiology, Yamagata University School of Medicine, Yamagata, Japan
| | - Miyuki Unekawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Ruka Adachi
- Division of Brain Sciences, Keio University School of Medicine, Tokyo, Japan
| | - Mayumi Yamada
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Itaru Imayoshi
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.,Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshifumi Abe
- Division of Brain Sciences, Keio University School of Medicine, Tokyo, Japan
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Kenji F Tanaka
- Division of Brain Sciences, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
38
|
Natsubori A, Hirai S, Kwon S, Ono D, Deng F, Wan J, Miyazawa M, Kojima T, Okado H, Karashima A, Li Y, Tanaka KF, Honda M. Serotonergic neurons control cortical neuronal intracellular energy dynamics by modulating astrocyte-neuron lactate shuttle. iScience 2023; 26:105830. [PMID: 36713262 PMCID: PMC9881222 DOI: 10.1016/j.isci.2022.105830] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 08/15/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
The central serotonergic system has multiple roles in animal physiology and behavior, including sleep-wake control. However, its function in controlling brain energy metabolism according to the state of animals remains undetermined. Through in vivo monitoring of energy metabolites and signaling, we demonstrated that optogenetic activation of raphe serotonergic neurons increased cortical neuronal intracellular concentration of ATP, an indispensable cellular energy molecule, which was suppressed by inhibiting neuronal uptake of lactate derived from astrocytes. Raphe serotonergic neuronal activation induced cortical astrocytic Ca2+ and cAMP surges and increased extracellular lactate concentrations, suggesting the facilitation of lactate release from astrocytes. Furthermore, chemogenetic inhibition of raphe serotonergic neurons partly attenuated the increase in cortical neuronal intracellular ATP levels as arousal increased in mice. Serotonergic neuronal activation promoted an increase in cortical neuronal intracellular ATP levels, partly mediated by the facilitation of the astrocyte-neuron lactate shuttle, contributing to state-dependent optimization of neuronal intracellular energy levels.
Collapse
Affiliation(s)
- Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan,Corresponding author
| | - Shinobu Hirai
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Soojin Kwon
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Daisuke Ono
- Department of Neuroscience Ⅱ, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan,Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Fei Deng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Jinxia Wan
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Momoka Miyazawa
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan,Faculty of Science Division Ⅱ, Tokyo University of Science, Shinjuku-ku, Tokyo 162-8601, Japan
| | - Takashi Kojima
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Haruo Okado
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Akihiro Karashima
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, Sendai 982-8577, Japan
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China,PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Makoto Honda
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
39
|
Median raphe serotonergic neurons projecting to the interpeduncular nucleus control preference and aversion. Nat Commun 2022; 13:7708. [PMID: 36550097 PMCID: PMC9780347 DOI: 10.1038/s41467-022-35346-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Appropriate processing of reward and aversive information is essential for survival. Although a critical role of serotonergic neurons in the dorsal raphe nucleus (DRN) in reward processing has been shown, the lack of rewarding effects with selective serotonin reuptake inhibitors (SSRIs) implies the presence of a discrete serotonergic system playing an opposite role to the DRN in the processing of reward and aversive stimuli. Here, we demonstrated that serotonergic neurons in the median raphe nucleus (MRN) of mice process reward and aversive information in opposite directions to DRN serotonergic neurons. We further identified MRN serotonergic neurons, including those projecting to the interpeduncular nucleus (5-HTMRN→IPN), as a key mediator of reward and aversive stimuli. Moreover, 5-HT receptors, including 5-HT2A receptors in the interpeduncular nucleus, are involved in the aversive properties of MRN serotonergic neural activity. Our findings revealed an essential function of MRN serotonergic neurons, including 5-HTMRN→IPN, in the processing of reward and aversive stimuli.
Collapse
|
40
|
Hazra D, Yoshinaga S, Yoshida K, Takata N, Tanaka KF, Kubo KI, Nakajima K. Rhythmic activation of excitatory neurons in the mouse frontal cortex improves the prefrontal cortex-mediated cognitive function. Cereb Cortex 2022; 32:5243-5258. [PMID: 35136976 DOI: 10.1093/cercor/bhac011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/27/2022] Open
Abstract
The prefrontal cortex (PFC) plays essential roles in cognitive processes. Previous studies have suggested the layer and the cell type-specific activation for cognitive enhancement. However, the mechanism by which a temporal pattern of activation affects cognitive function remains to be elucidated. Here, we investigated whether the specific activation of excitatory neurons in the superficial layers mainly in the PFC according to a rhythmic or nonrhythmic pattern could modulate the cognitive functions of normal mice. We used a C128S mutant of channelrhodopsin 2, a step function opsin, and administered two light illumination patterns: (i) alternating pulses of blue and yellow light for rhythmic activation or (ii) pulsed blue light only for nonrhythmic activation. Behavioral analyses were performed to compare the behavioral consequences of these two neural activation patterns. The alternating blue and yellow light pulses, but not the pulsed blue light only, significantly improved spatial working memory and social recognition without affecting motor activity or the anxiety level. These results suggest that the rhythmic, but not the nonrhythmic, activation could enhance cognitive functions. This study indicates that not only the population of neurons that are activated but also the pattern of activation plays a crucial role in the cognitive enhancement.
Collapse
Affiliation(s)
- Debabrata Hazra
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Satoshi Yoshinaga
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan.,Department of Anatomy, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Keitaro Yoshida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Norio Takata
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan.,Department of Anatomy, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
41
|
Morizawa YM, Matsumoto M, Nakashima Y, Endo N, Aida T, Ishikane H, Beppu K, Moritoh S, Inada H, Osumi N, Shigetomi E, Koizumi S, Yang G, Hirai H, Tanaka K, Tanaka KF, Ohno N, Fukazawa Y, Matsui K. Synaptic pruning through glial synapse engulfment upon motor learning. Nat Neurosci 2022; 25:1458-1469. [PMID: 36319770 DOI: 10.1038/s41593-022-01184-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 09/15/2022] [Indexed: 11/06/2022]
Abstract
Synaptic pruning is a fundamental process of neuronal circuit refinement in learning and memory. Accumulating evidence suggests that glia participates in sculpting the neuronal circuits through synapse engulfment. However, whether glial involvement in synaptic pruning has a role in memory formation remains elusive. Using newly developed phagocytosis reporter mice and three-dimensional ultrastructural characterization, we found that synaptic engulfment by cerebellar Bergmann glia (BG) frequently occurred upon cerebellum-dependent motor learning in mice. We observed increases in pre- and postsynaptic nibbling by BG along with a reduction in spine volume after learning. Pharmacological blockade of engulfment with Annexin V inhibited both the spine volume reduction and overnight improvement of motor adaptation. These results indicate that BG contribute to the refinement of the mature cerebellar cortical circuit through synaptic engulfment during motor learning.
Collapse
Affiliation(s)
- Yosuke M Morizawa
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan.
- JSPS Research Fellowship for Young Scientists, Tokyo, Japan.
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Mami Matsumoto
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Sciences, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Yuka Nakashima
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Narumi Endo
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Tomomi Aida
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroshi Ishikane
- Department of Psychology, Graduate School of Humanities, Senshu University, Kawasaki, Japan
| | - Kaoru Beppu
- Division of Interdisciplinary Medical Science, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Satoru Moritoh
- Department of Ophthalmology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hitoshi Inada
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Guang Yang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhiko Ohno
- Division of Histology and Cell Biology, Department of Anatomy, School of Medicine, Jichi Medical University, Tochigi, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Life Science Innovation Center, School of Medical Science, University of Fukui, Fukui, Japan
| | - Ko Matsui
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan.
- Division of Interdisciplinary Medical Science, Graduate School of Medicine, Tohoku University, Sendai, Japan.
| |
Collapse
|
42
|
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
|
43
|
Chemogenetic and Optogenetic Manipulations of Microglia in Chronic Pain. Neurosci Bull 2022; 39:368-378. [PMID: 35976535 PMCID: PMC10043090 DOI: 10.1007/s12264-022-00937-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/03/2022] [Indexed: 10/15/2022] Open
Abstract
Chronic pain relief remains an unmet medical need. Current research points to a substantial contribution of glia-neuron interaction in its pathogenesis. Particularly, microglia play a crucial role in the development of chronic pain. To better understand the microglial contribution to chronic pain, specific regional and temporal manipulations of microglia are necessary. Recently, two new approaches have emerged that meet these demands. Chemogenetic tools allow the expression of designer receptors exclusively activated by designer drugs (DREADDs) specifically in microglia. Similarly, optogenetic tools allow for microglial manipulation via the activation of artificially expressed, light-sensitive proteins. Chemo- and optogenetic manipulations of microglia in vivo are powerful in interrogating microglial function in chronic pain. This review summarizes these emerging tools in studying the role of microglia in chronic pain and highlights their potential applications in microglia-related neurological disorders.
Collapse
|
44
|
Takahashi A, Durand-de Cuttoli R, Flanigan ME, Hasegawa E, Tsunematsu T, Aleyasin H, Cherasse Y, Miya K, Okada T, Keino-Masu K, Mitsui K, Li L, Patel V, Blitzer RD, Lazarus M, Tanaka KF, Yamanaka A, Sakurai T, Ogawa S, Russo SJ. Lateral habenula glutamatergic neurons projecting to the dorsal raphe nucleus promote aggressive arousal in mice. Nat Commun 2022; 13:4039. [PMID: 35864121 PMCID: PMC9304121 DOI: 10.1038/s41467-022-31728-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
The dorsal raphe nucleus (DRN) is known to control aggressive behavior in mice. Here, we found that glutamatergic projections from the lateral habenula (LHb) to the DRN were activated in male mice that experienced pre-exposure to a rival male mouse ("social instigation") resulting in heightened intermale aggression. Both chemogenetic and optogenetic suppression of the LHb-DRN projection blocked heightened aggression after social instigation in male mice. In contrast, inhibition of this pathway did not affect basal levels of aggressive behavior, suggesting that the activity of the LHb-DRN projection is not necessary for the expression of species-typical aggressive behavior, but required for the increase of aggressive behavior resulting from social instigation. Anatomical analysis showed that LHb neurons synapse on non-serotonergic DRN neurons that project to the ventral tegmental area (VTA), and optogenetic activation of the DRN-VTA projection increased aggressive behaviors. Our results demonstrate that the LHb glutamatergic inputs to the DRN promote aggressive arousal induced by social instigation, which contributes to aggressive behavior by activating VTA-projecting non-serotonergic DRN neurons as one of its potential targets.
Collapse
Affiliation(s)
- Aki Takahashi
- Laboratory of Behavioral Neurobiology, Faculty of Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
- Nash Family Department of Neuroscience and Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Romain Durand-de Cuttoli
- Nash Family Department of Neuroscience and Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Meghan E Flanigan
- Nash Family Department of Neuroscience and Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, 27599, NC, USA
| | - Emi Hasegawa
- Department of Molecular Behavioral Physiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Tomomi Tsunematsu
- Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan
- Advanced Interdisciplinary Research Division, Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama, 332-0012, Japan
| | - Hossein Aleyasin
- Nash Family Department of Neuroscience and Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Ken Miya
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Takuya Okada
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kazuko Keino-Masu
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Koshiro Mitsui
- Laboratory of Behavioral Neurobiology, Faculty of Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Long Li
- Nash Family Department of Neuroscience and Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vishwendra Patel
- Department of Pharmacological Sciences and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert D Blitzer
- Department of Pharmacological Sciences and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Takeshi Sakurai
- Department of Molecular Behavioral Physiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan
| | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan
| | - Scott J Russo
- Nash Family Department of Neuroscience and Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
45
|
Rahaman SM, Chowdhury S, Mukai Y, Ono D, Yamaguchi H, Yamanaka A. Functional Interaction Between GABAergic Neurons in the Ventral Tegmental Area and Serotonergic Neurons in the Dorsal Raphe Nucleus. Front Neurosci 2022; 16:877054. [PMID: 35663550 PMCID: PMC9160575 DOI: 10.3389/fnins.2022.877054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/21/2022] [Indexed: 12/02/2022] Open
Abstract
GABAergic neurons in the ventral tegmental area (VTA) have brain-wide projections and are involved in multiple behavioral and physiological functions. Here, we revealed the responsiveness of Gad67+ neurons in VTA (VTAGad67+) to various neurotransmitters involved in the regulation of sleep/wakefulness by slice patch clamp recording. Among the substances tested, a cholinergic agonist activated, but serotonin, dopamine and histamine inhibited these neurons. Dense VTAGad67+ neuronal projections were observed in brain areas regulating sleep/wakefulness, including the central amygdala (CeA), dorsal raphe nucleus (DRN), and locus coeruleus (LC). Using a combination of electrophysiology and optogenetic studies, we showed that VTAGad67+ neurons inhibited all neurons recorded in the DRN, but did not inhibit randomly recorded neurons in the CeA and LC. Further examination revealed that the serotonergic neurons in the DRN (DRN5–HT) were monosynaptically innervated and inhibited by VTAGad67+ neurons. All recorded DRN5–HT neurons received inhibitory input from VTAGad67+ neurons, while only one quarter of them received inhibitory input from local GABAergic neurons. Gad67+ neurons in the DRN (DRNGad67+) also received monosynaptic inhibitory input from VTAGad67+ neurons. Taken together, we found that VTAGad67+ neurons were integrated in many inputs, and their output inhibits DRN5–HT neurons, which may regulate physiological functions including sleep/wakefulness.
Collapse
Affiliation(s)
- Sheikh Mizanur Rahaman
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Srikanta Chowdhury
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Yasutaka Mukai
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Yamaguchi
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- *Correspondence: Akihiro Yamanaka,
| |
Collapse
|
46
|
Dorgans K, Guo D, Kurima K, Wickens J, Uusisaari MY. Designing AAV Vectors for Monitoring the Subtle Calcium Fluctuations of Inferior Olive Network in vivo. Front Cell Neurosci 2022; 16:825056. [PMID: 35573836 PMCID: PMC9093741 DOI: 10.3389/fncel.2022.825056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Adeno-associated viral (AAV) vectors, used as vehicles for gene transfer into the brain, are a versatile and powerful tool of modern neuroscience that allow identifying specific neuronal populations, monitoring and modulating their activity. For consistent and reproducible results, the AAV vectors must be engineered so that they reliably and accurately target cell populations. Furthermore, transgene expression must be adjusted to sufficient and safe levels compatible with the physiology of studied cells. We undertook the effort to identify and validate an AAV vector that could be utilized for researching the inferior olivary (IO) nucleus, a structure gating critical timing-related signals to the cerebellum. By means of systematic construct generation and quantitative expression profiling, we succeeded in creating a viral tool for specific and strong transfection of the IO neurons without adverse effects on their physiology. The potential of these tools is demonstrated by expressing the calcium sensor GCaMP6s in adult mouse IO neurons. We could monitor subtle calcium fluctuations underlying two signatures of intrinsic IO activity: the subthreshold oscillations (STOs) and the variable-duration action potential waveforms both in-vitro and in-vivo. Further, we show that the expression levels of GCaMP6s allowing such recordings are compatible with the delicate calcium-based dynamics of IO neurons, inviting future work into the network dynamics of the olivo-cerebellar system in behaving animals.
Collapse
Affiliation(s)
- Kevin Dorgans
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Da Guo
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Kiyoto Kurima
- Neurobiology Research Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Jeff Wickens
- Neurobiology Research Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Marylka Yoe Uusisaari
- Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- *Correspondence: Marylka Yoe Uusisaari
| |
Collapse
|
47
|
Balachandar L, Borrego D, Diaz JR. Serotype-based evaluation of an optogenetic construct in rat cortical astrocytes. Biochem Biophys Res Commun 2022; 593:35-39. [DOI: 10.1016/j.bbrc.2022.01.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/08/2022] [Indexed: 11/16/2022]
|
48
|
Danjo Y, Shinozaki Y, Natsubori A, Kubota Y, Kashiwagi K, Tanaka KF, Koizumi S. The Mlc1 Promoter Directs Müller Cell-specific Gene Expression in the Retina. Transl Vis Sci Technol 2022; 11:25. [PMID: 35040915 PMCID: PMC8764212 DOI: 10.1167/tvst.11.1.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Because the importance of glia in regulating brain functions has been demonstrated, genetic technologies that manipulate glial cell-specific gene expression in the brain have become essential and have made great progress. However, it is unknown whether the same strategy that is used in the brain can be applied to the retina because retinal glia differs from glia in the brain. Here, we aimed to find a method for selective gene expression in Müller cells (characteristic glial cells in the retina) and identified Mlc1 as a specific promoter of Müller cells. Methods Mlc1-tTA::Yellow-Cameleon-NanotetO/tetO (YC-Nano) mice were used as a reporter line. YC-Nano, a fluorescent protein, was ectopically expressed in the cell type controlled by the Mlc1 promotor. Immunofluorescence staining was used to identify the cell type expressing YC-Nano protein. Results YC-Nano-positive (+) signals were observed as vertical stalks in the sliced retina and spanned from the nerve fiber layer through the outer nuclear layer. The density of YC-Nano+ cells was higher around the optic nerve head and lower in the peripheral retina. The YC-Nano+ signals colocalized with vimentin, a marker of Müller cells, but not with the cell markers for blood vessels, microglia, neurons, or astrocytes. Conclusions The Mlc1 promoter allows us to manipulate gene expression in Müller cells without affecting astrocytes in the retina. Translational Relevance Gene manipulation under control of Mlc1 promoter offers novel technique to investigate the role of Müller cells.
Collapse
Affiliation(s)
- Yosuke Danjo
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Akiyo Natsubori
- Sleep Disorders Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yuto Kubota
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Kenji Kashiwagi
- Department of Ophthalmology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
49
|
Shimoda Y, Beppu K, Ikoma Y, Morizawa YM, Zuguchi S, Hino U, Yano R, Sugiura Y, Moritoh S, Fukazawa Y, Suematsu M, Mushiake H, Nakasato N, Iwasaki M, Tanaka KF, Tominaga T, Matsui K. Optogenetic stimulus-triggered acquisition of seizure resistance. Neurobiol Dis 2021; 163:105602. [PMID: 34954320 DOI: 10.1016/j.nbd.2021.105602] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 11/26/2022] Open
Abstract
Unlike an electrical circuit, the hardware of the brain is susceptible to change. Repeated electrical brain stimulation mimics epileptogenesis. After such "kindling" process, a moderate stimulus would become sufficient in triggering a severe seizure. Here, we report that optogenetic neuronal stimulation can also convert the rat brain to a hyperexcitable state. However, continued stimulation once again converted the brain to a state that was strongly resistant to seizure induction. Histochemical examinations showed that moderate astrocyte activation was coincident with resilience acquisition. Administration of an adenosine A1 receptor antagonist instantly reverted the brain back to a hyperexcitable state, suggesting that hyperexcitability was suppressed by adenosine. Furthermore, an increase in basal adenosine was confirmed using in vivo microdialysis. Daily neuron-to-astrocyte signaling likely prompted a homeostatic increase in the endogenous actions of adenosine. Our data suggest that a certain stimulation paradigm could convert the brain circuit resilient to epilepsy without exogenous drug administration.
Collapse
Affiliation(s)
- Yoshiteru Shimoda
- Division of Interdisciplinary Medical Science, Center for Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Kaoru Beppu
- Division of Interdisciplinary Medical Science, Center for Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yoko Ikoma
- Super-network Brain Physiology, Tohoku University Graduate School of Life Sciences, Sendai 980-8577, Japan
| | - Yosuke M Morizawa
- Division of Interdisciplinary Medical Science, Center for Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; Super-network Brain Physiology, Tohoku University Graduate School of Life Sciences, Sendai 980-8577, Japan
| | - Satoshi Zuguchi
- Division of Interdisciplinary Medical Science, Center for Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Utaro Hino
- Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Ryutaro Yano
- Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Yuki Sugiura
- Department of Biochemistry & Integrative Medical Biology, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Satoru Moritoh
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yugo Fukazawa
- Division of Cell Biology and Neuroscience, University of Fukui Faculty of Medical Sciences, Fukui 910-1193, Japan
| | - Makoto Suematsu
- Department of Biochemistry & Integrative Medical Biology, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Hajime Mushiake
- Department of Physiology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Nobukazu Nakasato
- Department of Epileptology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Masaki Iwasaki
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Ko Matsui
- Division of Interdisciplinary Medical Science, Center for Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; Super-network Brain Physiology, Tohoku University Graduate School of Life Sciences, Sendai 980-8577, Japan.
| |
Collapse
|
50
|
Hatakeyama N, Unekawa M, Murata J, Tomita Y, Suzuki N, Nakahara J, Takuwa H, Kanno I, Matsui K, Tanaka KF, Masamoto K. Differential pial and penetrating arterial responses examined by optogenetic activation of astrocytes and neurons. J Cereb Blood Flow Metab 2021; 41:2676-2689. [PMID: 33899558 PMCID: PMC8504944 DOI: 10.1177/0271678x211010355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A variety of brain cells participates in neurovascular coupling by transmitting and modulating vasoactive signals. The present study aimed to probe cell type-dependent cerebrovascular (i.e., pial and penetrating arterial) responses with optogenetics in the cortex of anesthetized mice. Two lines of the transgenic mice expressing a step function type of light-gated cation channel (channelrhodopsine-2; ChR2) in either cortical neurons (muscarinic acetylcholine receptors) or astrocytes (Mlc1-positive) were used in the experiments. Photo-activation of ChR2-expressing astrocytes resulted in a widespread increase in cerebral blood flow (CBF), extending to the nonstimulated periphery. In contrast, photo-activation of ChR2-expressing neurons led to a relatively localized increase in CBF. The differences in the spatial extent of the CBF responses are potentially explained by differences in the involvement of the vascular compartments. In vivo imaging of the cerebrovascular responses revealed that ChR2-expressing astrocyte activation led to the dilation of both pial and penetrating arteries, whereas ChR2-expressing neuron activation predominantly caused dilation of the penetrating arterioles. Pharmacological studies showed that cell type-specific signaling mechanisms participate in the optogenetically induced cerebrovascular responses. In conclusion, pial and penetrating arterial vasodilation were differentially evoked by ChR2-expressing astrocytes and neurons.
Collapse
Affiliation(s)
- Nao Hatakeyama
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
| | - Miyuki Unekawa
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Juri Murata
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan
| | - Yutaka Tomita
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan.,Tomita Hospital, Aichi, Japan
| | - Norihiro Suzuki
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan.,Shonan Keiiku Hospital, Kanagawa, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Takuwa
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, Chiba, Japan
| | - Iwao Kanno
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, Chiba, Japan
| | - Ko Matsui
- Super-Network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Miyagi, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kazuto Masamoto
- Graduate School of Informatics and Engineering, University of Electro-Communications, Tokyo, Japan.,Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, Chiba, Japan.,Center for Neuroscience and Biomedical Engineering, University of Electro-Communications, Tokyo, Japan
| |
Collapse
|