1
|
Alexander SN, Reed OA, Burton MD. Spinal cord microglia drive sex differences in ethanol-mediated PGE2-induced allodynia. Brain Behav Immun 2024; 122:399-421. [PMID: 39147173 DOI: 10.1016/j.bbi.2024.08.026] [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: 02/02/2024] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024] Open
Abstract
The mechanisms of how long-term alcohol use can lead to persistent pain pathology are unclear. Understanding how earlier events of short-term alcohol use can lower the threshold of non-painful stimuli, described as allodynia could prove prudent to understand important initiating mechanisms. Previously, we observed that short-term low-dose alcohol intake induced female-specific allodynia and increased microglial activation in the spinal cord dorsal horn. Other literature describes how chronic ethanol exposure activates Toll-like receptor 4 (TLR4) to initiate inflammatory responses. TLR4 is expressed on many cell types, and we aimed to investigate whether TLR4 on microglia is sufficient to potentiate allodynia during a short-term/low-dose alcohol paradigm. Our study used a novel genetic model where TLR4 expression is removed from the entire body by introducing a floxed transcriptional blocker (TLR4-null background (TLR4LoxTB)), then restricted to microglia by breeding TLR4LoxTB animals with Cx3CR1:CreERT2 animals. As previously reported, after 14 days of ethanol administration alone, we observed no increased pain behavior. However, we observed significant priming effects 3 hrs post intraplantar injection of a subthreshold dose of prostaglandin E2 (PGE2) in wild-type and microglia-TLR4 restricted female mice. We also observed a significant female-specific shift to pro-inflammatory phenotype and morphological changes in microglia of the lumbar dorsal horn. Investigations in pain priming-associated neuronal subtypes showed an increase of c-Fos and FosB activity in PKCγ interneurons in the dorsal horn of female mice directly corresponding to increased microglial activity. This study uncovers cell- and female-specific roles of TLR4 in sexual dimorphisms in pain induction among non-pathological drinkers.
Collapse
Affiliation(s)
- Shevon N Alexander
- Neuroimmunology and Behavior Lab (NIB), Department of Neuroscience, School of Behavioral and Brain Science, Center for Advanced Pain Studies (CAPS), University of Texas at Dallas, Richardson, TX, USA
| | - Olivia A Reed
- Neuroimmunology and Behavior Lab (NIB), Department of Neuroscience, School of Behavioral and Brain Science, Center for Advanced Pain Studies (CAPS), University of Texas at Dallas, Richardson, TX, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Lab (NIB), Department of Neuroscience, School of Behavioral and Brain Science, Center for Advanced Pain Studies (CAPS), University of Texas at Dallas, Richardson, TX, USA.
| |
Collapse
|
2
|
Nevelchuk S, Brawek B, Schwarz N, Valiente-Gabioud A, Wuttke TV, Kovalchuk Y, Koch H, Höllig A, Steiner F, Figarella K, Griesbeck O, Garaschuk O. Morphotype-specific calcium signaling in human microglia. J Neuroinflammation 2024; 21:175. [PMID: 39020359 PMCID: PMC11256502 DOI: 10.1186/s12974-024-03169-6] [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: 05/25/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Key functions of Ca2+ signaling in rodent microglia include monitoring the brain state as well as the surrounding neuronal activity and sensing the danger or damage in their vicinity. Microglial Ca2+ dyshomeostasis is a disease hallmark in many mouse models of neurological disorders but the Ca2+ signal properties of human microglia remain unknown. METHODS We developed a novel genetically-encoded ratiometric Ca2+ indicator, targeting microglial cells in the freshly resected human tissue, organotypically cultured tissue slices and analyzed in situ ongoing Ca2+ signaling of decades-old microglia dwelling in their native microenvironment. RESULTS The data revealed marked compartmentalization of Ca2+ signals, with signal properties differing across the compartments and resident morphotypes. The basal Ca2+ levels were low in ramified and high in ameboid microglia. The fraction of cells with ongoing Ca2+ signaling, the fraction and the amplitude of process Ca2+ signals and the duration of somatic Ca2+ signals decreased when moving from ramified via hypertrophic to ameboid microglia. In contrast, the size of active compartments, the fraction and amplitude of somatic Ca2+ signals and the duration of process Ca2+ signals increased along this pathway.
Collapse
Affiliation(s)
- Sofia Nevelchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Bianca Brawek
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Niklas Schwarz
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ariel Valiente-Gabioud
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Thomas V Wuttke
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Department of Neurosurgery, University of Tübingen, Tübingen, Germany
| | - Yury Kovalchuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Henner Koch
- Department of Epileptology, Neurology, RWTH Aachen University Hospital, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, RWTH Aachen University, Aachen, Germany
| | - Frederik Steiner
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
| | - Katherine Figarella
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Oliver Griesbeck
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Keplerstr. 15, 72074, Tübingen, Germany.
| |
Collapse
|
3
|
Umpierre AD, Li B, Ayasoufi K, Simon WL, Zhao S, Xie M, Thyen G, Hur B, Zheng J, Liang Y, Bosco DB, Maynes MA, Wu Z, Yu X, Sung J, Johnson AJ, Li Y, Wu LJ. Microglial P2Y 6 calcium signaling promotes phagocytosis and shapes neuroimmune responses in epileptogenesis. Neuron 2024; 112:1959-1977.e10. [PMID: 38614103 PMCID: PMC11189754 DOI: 10.1016/j.neuron.2024.03.017] [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: 06/29/2023] [Revised: 01/09/2024] [Accepted: 03/13/2024] [Indexed: 04/15/2024]
Abstract
Microglial calcium signaling is rare in a baseline state but strongly engaged during early epilepsy development. The mechanism(s) governing microglial calcium signaling are not known. By developing an in vivo uridine diphosphate (UDP) fluorescent sensor, GRABUDP1.0, we discovered that UDP release is a conserved response to seizures and excitotoxicity across brain regions. UDP can signal through the microglial-enriched P2Y6 receptor to increase calcium activity during epileptogenesis. P2Y6 calcium activity is associated with lysosome biogenesis and enhanced production of NF-κB-related cytokines. In the hippocampus, knockout of the P2Y6 receptor prevents microglia from fully engulfing neurons. Attenuating microglial calcium signaling through calcium extruder ("CalEx") expression recapitulates multiple features of P2Y6 knockout, including reduced lysosome biogenesis and phagocytic interactions. Ultimately, P2Y6 knockout mice retain more CA3 neurons and better cognitive task performance during epileptogenesis. Our results demonstrate that P2Y6 signaling impacts multiple aspects of myeloid cell immune function during epileptogenesis.
Collapse
Affiliation(s)
| | - Bohan Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | | | - Whitney L Simon
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Grace Thyen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Benjamin Hur
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yue Liang
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mark A Maynes
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
| | - Xinzhu Yu
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jaeyun Sung
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA; Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Aaron J Johnson
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA; Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Nagarajan N, Capecchi MR. Optogenetic stimulation of mouse Hoxb8 microglia in specific regions of the brain induces anxiety, grooming, or both. Mol Psychiatry 2024; 29:1726-1740. [PMID: 37037872 PMCID: PMC11371632 DOI: 10.1038/s41380-023-02019-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 02/17/2023] [Accepted: 02/24/2023] [Indexed: 04/12/2023]
Abstract
Previously, we have shown that either disruption of the Hoxb8 gene or ablation of a microglial subpopulation, Hoxb8 microglia, results in mice exhibiting both chronic anxiety and OCSD-like behavior, compulsive pathological hair pulling (trichotillomania), to the point of showing lesions at the sites of overgrooming. Herein we show, that optogenetic stimulation of Hoxb8 microglia in specific regions of the brain induces elevated anxiety, grooming or both. Optogenetic stimulation of Hoxb8 microglia within the dorsomedial striatum (DMS) or the medial prefrontal cortex (mPFC) induces grooming, whereas stimulation of Hoxb8 microglia in the basolateral amygdala (BLA) or central amygdala (CeA) produces elevated anxiety. Optogenetic stimulation of Hoxb8 microglia in the ventral CA1 region of the hippocampus (vCA1) induces both behaviors as well as freezing. In vitro we directly demonstrate that optogenetic stimulation of Hoxb8 microglia in specific regions of the brain activate neighboring neural activity through the induction of the c-fos-immediate early response. These experiments connect outputs from optogenetically stimulated Hoxb8 microglia, within specific regions of the brain, to the activation of neurons and neural circuits that in turn enable induction of these behaviors. These experiments suggest that Hoxb8 microglia are likely to be among, or the main, first responders to signals that evoke these behaviors. The same regions of the brain (DMS, mPFC, BLA, CeA and vCA1) have previously been defined at the neuronal level, by optogenetics, to control anxiety in mice. Intriguingly, the optogenetic experiments in microglia suggest that the two populations of microglia, canonical non-Hoxb8 and Hoxb8 microglia, function in opposition rather than in parallel to each other, providing a biological reason for the presence of two microglial subpopulations in mice.
Collapse
Affiliation(s)
- Naveen Nagarajan
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA.
| | - Mario R Capecchi
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA.
| |
Collapse
|
5
|
Wallis GJ, Bell LA, Wagner JN, Buxton L, Balachandar L, Wilcox KS. Reactive microglia fail to respond to environmental damage signals in a viral-induced mouse model of temporal lobe epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583768. [PMID: 38558969 PMCID: PMC10979929 DOI: 10.1101/2024.03.06.583768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Microglia are highly adaptable innate immune cells that rapidly respond to damage signals in the brain through adoption of a reactive phenotype and production of defensive inflammatory cytokines. Microglia express a distinct transcriptome, encoding receptors that allow them to dynamically respond to pathogens, damage signals, and cellular debris. Expression of one such receptor, the microglia-specific purinergic receptor P2ry12, is known to be downregulated in reactive microglia. Here, we explore the microglial response to purinergic damage signals in reactive microglia in the TMEV mouse model of viral brain infection and temporal lobe epilepsy. Using two-photon calcium imaging in acute hippocampal brain slices, we found that the ability of microglia to detect damage signals, engage calcium signaling pathways, and chemoattract towards laser-induced tissue damage was dramatically reduced during the peak period of seizures, cytokine production, and infection. Using combined RNAscope in situ hybridization and immunohistochemistry, we found that during this same stage of heightened infection and seizures, microglial P2ry12 expression was reduced, while the pro-inflammatory cytokine TNF-a expression was upregulated in microglia, suggesting that the depressed ability of microglia to respond to new damage signals via P2ry12 occurs during the time when local elevated cytokine production contributes to seizure generation following infection. Therefore, changes in microglial purinergic receptors during infection likely limit the ability of reactive microglia to respond to new threats in the CNS and locally contain the scale of the innate immune response in the brain.
Collapse
Affiliation(s)
- Glenna J. Wallis
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
| | - Laura A. Bell
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, 80904
| | - John N. Wagner
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
| | - Lauren Buxton
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
| | - Lakshmini Balachandar
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
| | - Karen S. Wilcox
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 80904
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, 80904
| |
Collapse
|
6
|
Saito K, Shigetomi E, Shinozaki Y, Kobayashi K, Parajuli B, Kubota Y, Sakai K, Miyakawa M, Horiuchi H, Nabekura J, Koizumi S. Microglia sense astrocyte dysfunction and prevent disease progression in an Alexander disease model. Brain 2024; 147:698-716. [PMID: 37955589 PMCID: PMC10834242 DOI: 10.1093/brain/awad358] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023] Open
Abstract
Alexander disease (AxD) is an intractable neurodegenerative disorder caused by GFAP mutations. It is a primary astrocyte disease with a pathological hallmark of Rosenthal fibres within astrocytes. AxD astrocytes show several abnormal phenotypes. Our previous study showed that AxD astrocytes in model mice exhibit aberrant Ca2+ signals that induce AxD aetiology. Here, we show that microglia have unique phenotypes with morphological and functional alterations, which are related to the pathogenesis of AxD. Immunohistochemical studies of 60TM mice (AxD model) showed that AxD microglia exhibited highly ramified morphology. Functional changes in microglia were assessed by Ca2+ imaging using hippocampal brain slices from Iba1-GCaMP6-60TM mice and two-photon microscopy. We found that AxD microglia showed aberrant Ca2+ signals, with high frequency Ca2+ signals in both the processes and cell bodies. These microglial Ca2+ signals were inhibited by pharmacological blockade or genetic knockdown of P2Y12 receptors but not by tetrodotoxin, indicating that these signals are independent of neuronal activity but dependent on extracellular ATP from non-neuronal cells. Our single-cell RNA sequencing data showed that the expression level of Entpd2, an astrocyte-specific gene encoding the ATP-degrading enzyme NTPDase2, was lower in AxD astrocytes than in wild-type astrocytes. In situ ATP imaging using the adeno-associated virus vector GfaABC1D ATP1.0 showed that exogenously applied ATP was present longer in 60TM mice than in wild-type mice. Thus, the increased ATP level caused by the decrease in its metabolizing enzyme in astrocytes could be responsible for the enhancement of microglial Ca2+ signals. To determine whether these P2Y12 receptor-mediated Ca2+ signals in AxD microglia play a significant role in the pathological mechanism, a P2Y12 receptor antagonist, clopidogrel, was administered. Clopidogrel significantly exacerbated pathological markers in AxD model mice and attenuated the morphological features of microglia, suggesting that microglia play a protective role against AxD pathology via P2Y12 receptors. Taken together, we demonstrated that microglia sense AxD astrocyte dysfunction via P2Y12 receptors as an increase in extracellular ATP and alter their morphology and Ca2+ signalling, thereby protecting against AxD pathology. Although AxD is a primary astrocyte disease, our study may facilitate understanding of the role of microglia as a disease modifier, which may contribute to the clinical diversity of AxD.
Collapse
Affiliation(s)
- Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kenji Kobayashi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Bijay Parajuli
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Yuto Kubota
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kent Sakai
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Miho Miyakawa
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Hiroshi Horiuchi
- Division of Homeostatic Development, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585, Japan
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
7
|
Izquierdo P, Jolivet RB, Attwell D, Madry C. Amyloid plaques and normal ageing have differential effects on microglial Ca 2+ activity in the mouse brain. Pflugers Arch 2024; 476:257-270. [PMID: 37966547 PMCID: PMC10791787 DOI: 10.1007/s00424-023-02871-3] [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: 08/10/2023] [Revised: 10/02/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023]
Abstract
In microglia, changes in intracellular calcium concentration ([Ca2+]i) may regulate process motility, inflammasome activation, and phagocytosis. However, while neurons and astrocytes exhibit frequent spontaneous Ca2+ activity, microglial Ca2+ signals are much rarer and poorly understood. Here, we studied [Ca2+]i changes of microglia in acute brain slices using Fluo-4-loaded cells and mice expressing GCaMP5g in microglia. Spontaneous Ca2+ transients occurred ~ 5 times more frequently in individual microglial processes than in their somata. We assessed whether microglial Ca2+ responses change in Alzheimer's disease (AD) using AppNL-G-F knock-in mice. Proximity to Aβ plaques strongly affected microglial Ca2+ activity. Although spontaneous Ca2+ transients were unaffected in microglial processes, they were fivefold more frequent in microglial somata near Aβ plaques than in wild-type microglia. Microglia away from Aβ plaques in AD mice showed intermediate properties for morphology and Ca2+ responses, partly resembling those of wild-type microglia. By contrast, somatic Ca2+ responses evoked by tissue damage were less intense in microglia near Aβ plaques than in wild-type microglia, suggesting different mechanisms underlying spontaneous vs. damage-evoked Ca2+ signals. Finally, as similar processes occur in neurodegeneration and old age, we studied whether ageing affected microglial [Ca2+]i. Somatic damage-evoked Ca2+ responses were greatly reduced in microglia from old mice, as in the AD mice. In contrast to AD, however, old age did not alter the occurrence of spontaneous Ca2+ signals in microglial somata but reduced the rate of events in processes. Thus, we demonstrate distinct compartmentalised Ca2+ activity in microglia from healthy, aged and AD-like brains.
Collapse
Affiliation(s)
- Pablo Izquierdo
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| | - Renaud B Jolivet
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Paul-Henri Spaaklaan 1, 6229 EN, Maastricht, The Netherlands
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK.
| | - Christian Madry
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK.
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Institute of Neurophysiology, 10117, Berlin, Germany.
| |
Collapse
|
8
|
Crockett A, Fuhrmann M, Garaschuk O, Davalos D. Progress in Structural and Functional In Vivo Imaging of Microglia and Their Application in Health and Disease. ADVANCES IN NEUROBIOLOGY 2024; 37:65-80. [PMID: 39207687 DOI: 10.1007/978-3-031-55529-9_5] [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: 09/04/2024]
Abstract
The first line of defense for the central nervous system (CNS) against injury or disease is provided by microglia. Microglia were long believed to stay in a dormant/resting state, reacting only to injury or disease. This view changed dramatically with the development of modern imaging techniques that allowed the study of microglial behavior in the intact brain over time, to reveal the dynamic nature of their responses. Over the past two decades, in vivo imaging using multiphoton microscopy has revealed numerous new functions of microglia in the developing, adult, aged, injured, and diseased CNS. As the most dynamic cells in the brain, microglia continuously contact all structures and cell types, such as glial and vascular cells, neuronal cell bodies, axons, dendrites, and dendritic spines, and are believed to play a central role in sculpting neuronal networks throughout life. Following trauma, or in neurodegenerative or neuroinflammatory diseases, microglial responses range from protective to harmful, underscoring the need to better understand their diverse roles and states in different pathological conditions. In this chapter, we introduce multiphoton microscopy and discuss recent advances in structural and functional imaging technologies that have expanded our toolbox to study microglial states and behaviors in new ways and depths. We also discuss relevant mouse models available for in vivo imaging studies of microglia and review how such studies are constantly refining our understanding of the multifaceted role of microglia in the healthy and diseased CNS.
Collapse
Affiliation(s)
- Alexis Crockett
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Martin Fuhrmann
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Olga Garaschuk
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Dimitrios Davalos
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
9
|
He Y, Liu T, He Q, Ke W, Li X, Du J, Deng S, Shu Z, Wu J, Yang B, Wang Y, Mao Y, Rao Y, Shu Y, Peng B. Microglia facilitate and stabilize the response to general anesthesia via modulating the neuronal network in a brain region-specific manner. eLife 2023; 12:RP92252. [PMID: 38131301 PMCID: PMC10746144 DOI: 10.7554/elife.92252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
General anesthesia leads to a loss of consciousness and an unrousable state in patients. Although general anesthetics are widely used in clinical practice, their underlying mechanisms remain elusive. The potential involvement of nonneuronal cells is unknown. Microglia are important immune cells in the central nervous system (CNS) that play critical roles in CNS function and dysfunction. We unintentionally observed delayed anesthesia induction and early anesthesia emergence in microglia-depleted mice. We found that microglial depletion differentially regulates neuronal activities by suppressing the neuronal network of anesthesia-activated brain regions and activating emergence-activated brain regions. Thus, microglia facilitate and stabilize the anesthesia status. This influence is not mediated by dendritic spine plasticity. Instead, it relies on the activation of microglial P2Y12 and subsequent calcium influx, which facilitates the general anesthesia response. Together, we elucidate the regulatory role of microglia in general anesthesia, extending our knowledge of how nonneuronal cells modulate neuronal activities.
Collapse
Affiliation(s)
- Yang He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Taohui Liu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Quansheng He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Wei Ke
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Xiaoyu Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jinjin Du
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Suixin Deng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Zhenfeng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jialin Wu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Baozhi Yang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Yuqing Wang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
| | - Yousheng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- Co-Innovation Center of Neurodegeneration, Nantong UniversityNantongChina
| |
Collapse
|
10
|
Pan K, Garaschuk O. The role of intracellular calcium-store-mediated calcium signals in in vivo sensor and effector functions of microglia. J Physiol 2023; 601:4203-4215. [PMID: 35315518 DOI: 10.1113/jp279521] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/09/2022] [Indexed: 11/08/2022] Open
Abstract
Under physiological conditions microglia, the immune sentinels of the brain, constantly monitor their microenvironment. In the case of danger, damage or cell/tissue dyshomeostasis, they react with changes in process motility, polarization, directed process movement, morphology and gene expression profile; release pro- and anti-inflammatory mediators; proliferate; and clean brain parenchyma by means of phagocytosis. Based on recent transcriptomic and in vivo Ca2+ imaging data, we argue that the local cell/tissue dyshomeostasis is sensed by microglia via intracellular Ca2+ signals, many of which are mediated by Ca2+ release from the intracellular Ca2+ stores. These signals encode the strength, duration and spatiotemporal pattern of the stimulus and, at the same time, relay this information further to trigger the respective Ca2+ -dependent effector pathways. We also point to the fact that microglial Ca2+ signalling is sexually dimorphic and undergoes profound changes across the organism's lifespan. Interestingly, the first changes in microglial Ca2+ signalling are visible already in 9- to 11-month-old mice, roughly corresponding to 40-year-old humans.
Collapse
Affiliation(s)
- Kuang Pan
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
11
|
Petry P, Oschwald A, Kierdorf K. Microglial tissue surveillance: The never-resting gardener in the developing and adult CNS. Eur J Immunol 2023; 53:e2250232. [PMID: 37042800 DOI: 10.1002/eji.202250232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/24/2023] [Accepted: 04/11/2023] [Indexed: 04/13/2023]
Abstract
Immunosurveillance by microglia is a dynamic process in the central nervous system (CNS) with versatile functions to maintain tissue homeostasis and provide immune defense. A tightly controlled microglia network throughout the CNS parenchyma facilitates efficient immunosurveillance, where each cell guards a certain tissue territory. Each cell is constantly surveilling its environment and the surrounding cells, screening for pathogens but also removing cell debris and metabolites, grooming neighboring cells and facilitating cellular crosstalk. In the absence of inflammation, this "tissue surveillance" by microglia presents an essential process for CNS homeostasis and development. In this review, we provide a summary on different tissue surveillance functions mediated by microglia, the underlying molecular machineries, and how defects, such as genetic mutations, can alter these surveillance mechanisms and cause disease onset.
Collapse
Affiliation(s)
- Philippe Petry
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Alexander Oschwald
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
12
|
Sokolowski JD, Soldozy S, Sharifi KA, Norat P, Kearns KN, Liu L, Williams AM, Yağmurlu K, Mastorakos P, Miller GW, Kalani MYS, Park MS, Kellogg RT, Tvrdik P. Preclinical models of middle cerebral artery occlusion: new imaging approaches to a classic technique. Front Neurol 2023; 14:1170675. [PMID: 37409019 PMCID: PMC10318149 DOI: 10.3389/fneur.2023.1170675] [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: 02/21/2023] [Accepted: 05/30/2023] [Indexed: 07/07/2023] Open
Abstract
Stroke remains a major burden on patients, families, and healthcare professionals, despite major advances in prevention, acute treatment, and rehabilitation. Preclinical basic research can help to better define mechanisms contributing to stroke pathology, and identify therapeutic interventions that can decrease ischemic injury and improve outcomes. Animal models play an essential role in this process, and mouse models are particularly well-suited due to their genetic accessibility and relatively low cost. Here, we review the focal cerebral ischemia models with an emphasis on the middle cerebral artery occlusion technique, a "gold standard" in surgical ischemic stroke models. Also, we highlight several histologic, genetic, and in vivo imaging approaches, including mouse stroke MRI techniques, that have the potential to enhance the rigor of preclinical stroke evaluation. Together, these efforts will pave the way for clinical interventions that can mitigate the negative impact of this devastating disease.
Collapse
Affiliation(s)
- Jennifer D. Sokolowski
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Sauson Soldozy
- Department of Neurological Surgery, Westchester Medical Center, Valhalla, NY, United States
| | - Khadijeh A. Sharifi
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Pedro Norat
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Kathryn N. Kearns
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Lei Liu
- Department of Neurological Surgery and Neuroscience, Northwestern University, Chicago, IL, United States
| | - Ashley M. Williams
- School of Medicine, Morsani College of Medicine, Tampa, FL, United States
| | - Kaan Yağmurlu
- Department of Neurological Surgery, University of Tennessee, Memphis, TN, United States
| | - Panagiotis Mastorakos
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - G. Wilson Miller
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, United States
| | - M. Yashar S. Kalani
- Department of Neurological Surgery, St. John's Neuroscience Institute, Tulsa, OK, United States
| | - Min S. Park
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Ryan T. Kellogg
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Petr Tvrdik
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
13
|
Umpierre AD, Li B, Ayasoufi K, Zhao S, Xie M, Thyen G, Hur B, Zheng J, Liang Y, Wu Z, Yu X, Sung J, Johnson AJ, Li Y, Wu LJ. Microglial P2Y 6 calcium signaling promotes phagocytosis and shapes neuroimmune responses in epileptogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544691. [PMID: 37398001 PMCID: PMC10312639 DOI: 10.1101/2023.06.12.544691] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Microglial calcium signaling is rare in a baseline state but shows strong engagement during early epilepsy development. The mechanism and purpose behind microglial calcium signaling is not known. By developing an in vivo UDP fluorescent sensor, GRABUDP1.0, we discovered that UDP release is a conserved response to seizures and excitotoxicity across brain regions. UDP signals to the microglial P2Y6 receptor for broad increases in calcium signaling during epileptogenesis. UDP-P2Y6 signaling is necessary for lysosome upregulation across limbic brain regions and enhances production of pro-inflammatory cytokines-TNFα and IL-1β. Failures in lysosome upregulation, observed in P2Y6 KO mice, can also be phenocopied by attenuating microglial calcium signaling in Calcium Extruder ("CalEx") mice. In the hippocampus, only microglia with P2Y6 expression can perform full neuronal engulfment, which substantially reduces CA3 neuron survival and impairs cognition. Our results demonstrate that calcium activity, driven by UDP-P2Y6 signaling, is a signature of phagocytic and pro-inflammatory function in microglia during epileptogenesis.
Collapse
Affiliation(s)
- Anthony D. Umpierre
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- These authors contributed equally
| | - Bohan Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Peking University School of Life Sciences, Beijing, CN 100871
- These authors contributed equally
| | | | - Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Neuroscience Track, Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905
| | - Manling Xie
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Neuroscience Track, Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905
| | - Grace Thyen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
| | - Benjamin Hur
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905
- Division of Surgery Research, Department of Surgery, Mayo Clinic, Rochester, MN 55905
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Neuroscience Track, Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905
| | - Yue Liang
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Peking University School of Life Sciences, Beijing, CN 100871
| | - Xinzhu Yu
- Department of Molecular and Integrative Physiology, Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Jaeyun Sung
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905
- Division of Surgery Research, Department of Surgery, Mayo Clinic, Rochester, MN 55905
| | - Aaron J. Johnson
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Immunology, Mayo Clinic, Rochester, MN 55905
- Department of Molecular Medicine, Mayo Clinic, Rochester MN 55905
| | - Yulong Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, Peking University School of Life Sciences, Beijing, CN 100871
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Immunology, Mayo Clinic, Rochester, MN 55905
- Lead contact
| |
Collapse
|
14
|
Caruso G, Di Pietro L, Caraci F. Gap Junctions and Connexins in Microglia-Related Oxidative Stress and Neuroinflammation: Perspectives for Drug Discovery. Biomolecules 2023; 13:biom13030505. [PMID: 36979440 PMCID: PMC10046203 DOI: 10.3390/biom13030505] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Microglia represent the immune system of the brain. Their role is central in two phenomena, neuroinflammation and oxidative stress, which are at the roots of different pathologies related to the central nervous system (CNS). In order to maintain the homeostasis of the brain and re-establish the equilibrium after a threatening imbalance, microglia communicate with each other and other cells within the CNS by receiving specific signals through membrane-bound receptors and then releasing neurotrophic factors into either the extracellular milieu or directly into the cytoplasm of nearby cells, such as astrocytes and neurons. These last two mechanisms rely on the activity of protein structures that enable the formation of channels in the membrane, namely, connexins and pannexins, that group and form gap junctions, hemichannels, and pannexons. These channels allow the release of gliotransmitters, such as adenosine triphosphate (ATP) and glutamate, together with calcium ion (Ca2+), that seem to play a pivotal role in inter-cellular communication. The aim of the present review is focused on the physiology of channel protein complexes and their contribution to neuroinflammatory and oxidative stress-related phenomena, which play a central role in neurodegenerative disorders. We will then discuss how pharmacological modulation of these channels can impact neuroinflammatory phenomena and hypothesize that currently available nutraceuticals, such as carnosine and N-acetylcysteine, can modulate the activity of connexins and pannexins in microglial cells and reduce oxidative stress in neurodegenerative disorders.
Collapse
Affiliation(s)
- Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, 95123 Catania, Italy
- Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
- Correspondence: ; Tel.: +39-0957385036
| | - Lucia Di Pietro
- Department of Drug and Health Sciences, University of Catania, 95123 Catania, Italy
- Scuola Superiore di Catania, University of Catania, 95123 Catania, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95123 Catania, Italy
- Unit of Neuropharmacology and Translational Neurosciences, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| |
Collapse
|
15
|
Jiang B, Ding T, Guo C, Bai X, Cao D, Wu X, Sha W, Jiang M, Wu L, Gao Y. NFAT1 Orchestrates Spinal Microglial Transcription and Promotes Microglial Proliferation via c-MYC Contributing to Nerve Injury-Induced Neuropathic Pain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201300. [PMID: 35892263 PMCID: PMC9507349 DOI: 10.1002/advs.202201300] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/17/2022] [Indexed: 05/17/2023]
Abstract
Peripheral nerve injury-induced spinal microglial proliferation plays a pivotal role in neuropathic pain. So far, key intracellular druggable molecules involved in this process are not identified. The nuclear factor of activated T-cells (NFAT1) is a master regulator of immune cell proliferation. Whether and how NFAT1 modulates spinal microglial proliferation during neuropathic pain remain unknown. Here it is reported that NFAT1 is persistently upregulated in microglia after spinal nerve ligation (SNL), which is regulated by TET2-mediated DNA demethylation. Global or microglia-specific deletion of Nfat1 attenuates SNL-induced pain and decreases excitatory synaptic transmission of lamina II neurons. Furthermore, deletion of Nfat1 decreases microglial proliferation and the expression of multiple microglia-related genes, such as cytokines, transmembrane signaling receptors, and transcription factors. Particularly, SNL increases the binding of NFAT1 with the promoter of Itgam, Tnf, Il-1b, and c-Myc in the spinal cord. Microglia-specific overexpression of c-MYC induces pain hypersensitivity and microglial proliferation. Finally, inhibiting NFAT1 and c-MYC by intrathecal injection of inhibitor or siRNA alleviates SNL-induced neuropathic pain. Collectively, NFAT1 is a hub transcription factor that regulates microglial proliferation via c-MYC and guides the expression of the activated microglia genome. Thus, NFAT1 may be an effective target for treating neuropathic pain.
Collapse
Affiliation(s)
- Bao‐Chun Jiang
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Ting‐Yu Ding
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Chang‐Yun Guo
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Xue‐Hui Bai
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - De‐Li Cao
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Xiao‐Bo Wu
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Wei‐Lin Sha
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Ming Jiang
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Long‐Jun Wu
- Department of NeurologyMayo ClinicRochesterMN55905USA
| | - Yong‐Jing Gao
- Institute of Pain Medicine and Special Environmental MedicineCo‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| |
Collapse
|
16
|
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
|
17
|
Wendimu MY, Hooks SB. Microglia Phenotypes in Aging and Neurodegenerative Diseases. Cells 2022; 11:2091. [PMID: 35805174 PMCID: PMC9266143 DOI: 10.3390/cells11132091] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/08/2023] Open
Abstract
Neuroinflammation is a hallmark of many neurodegenerative diseases (NDs) and plays a fundamental role in mediating the onset and progression of disease. Microglia, which function as first-line immune guardians of the central nervous system (CNS), are the central drivers of neuroinflammation. Numerous human postmortem studies and in vivo imaging analyses have shown chronically activated microglia in patients with various acute and chronic neuropathological diseases. While microglial activation is a common feature of many NDs, the exact role of microglia in various pathological states is complex and often contradictory. However, there is a consensus that microglia play a biphasic role in pathological conditions, with detrimental and protective phenotypes, and the overall response of microglia and the activation of different phenotypes depends on the nature and duration of the inflammatory insult, as well as the stage of disease development. This review provides a comprehensive overview of current research on the various microglia phenotypes and inflammatory responses in health, aging, and NDs, with a special emphasis on the heterogeneous phenotypic response of microglia in acute and chronic diseases such as hemorrhagic stroke (HS), Alzheimer's disease (AD), and Parkinson's disease (PD). The primary focus is translational research in preclinical animal models and bulk/single-cell transcriptome studies in human postmortem samples. Additionally, this review covers key microglial receptors and signaling pathways that are potential therapeutic targets to regulate microglial inflammatory responses during aging and in NDs. Additionally, age-, sex-, and species-specific microglial differences will be briefly reviewed.
Collapse
Affiliation(s)
| | - Shelley B. Hooks
- Hooks Lab, Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
18
|
Comparison of Microglial Morphology and Function in Primary Cerebellar Cell Cultures on Collagen and Collagen-Mimetic Hydrogels. Biomedicines 2022; 10:biomedicines10051023. [PMID: 35625762 PMCID: PMC9139096 DOI: 10.3390/biomedicines10051023] [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: 03/23/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 12/07/2022] Open
Abstract
Neuronal-glial cell cultures are usually grown attached to or encapsulated in an adhesive environment as evenly distributed networks lacking tissue-like cell density, organization and morphology. In such cultures, microglia have activated amoeboid morphology and do not display extended and intensively branched processes characteristic of the ramified tissue microglia. We have recently described self-assembling functional cerebellar organoids promoted by hydrogels containing collagen-like peptides (CLPs) conjugated to a polyethylene glycol (PEG) core. Spontaneous neuronal activity was accompanied by changes in the microglial morphology and behavior, suggesting the cells might play an essential role in forming the functional neuronal networks in response to the peptide signalling. The present study examines microglial cell morphology and function in cerebellar cell organoid cultures on CLP-PEG hydrogels and compares them to the cultures on crosslinked collagen hydrogels of similar elastomechanical properties. Material characterization suggested more expressed fibril orientation and denser packaging in crosslinked collagen than CLP-PEG. However, CLP-PEG promoted a significantly higher microglial motility (determined by time-lapse imaging) accompanied by highly diverse morphology including the ramified (brightfield and confocal microscopy), more active Ca2+ signalling (intracellular Ca2+ fluorescence recordings), and moderate inflammatory cytokine level (ELISA). On the contrary, on the collagen hydrogels, microglial cells were significantly less active and mostly round-shaped. In addition, the latter hydrogels did not support the neuron synaptic activity. Our findings indicate that the synthetic CLP-PEG hydrogels ensure more tissue-like microglial morphology, motility, and function than the crosslinked collagen substrates.
Collapse
|
19
|
Zhang H, Tan C, Shi X, Xu J. Impacts of autofluorescence on fluorescence based techniques to study microglia. BMC Neurosci 2022; 23:21. [PMID: 35361108 PMCID: PMC8973892 DOI: 10.1186/s12868-022-00703-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Microglia, the resident immune cells in the central nervous system, accrue autofluorescent granules inside their cytoplasm throughout their lifespan. In this report, we studied the impacts of autofluorescence on widely used fluorescence-based techniques to study microglia, including flow cytometry, immunofluorescence staining, and live imaging. RESULTS The failed attempt of using fluorescein isothiocyanate (FITC) conjugated antibody to detect lymphocyte-activation gene 3 protein in microglia prompted us to compare the sensitivity of FITC, phycoerythrin (PE) and allophycocyanin (APC) conjugated antibodies to detect surface protein expression in microglia. We found that PE outperformed FITC and APC as the fluorophore conjugated to antibody for flow cytometry by overcoming the interference from microglia autofluorescence. To identify the location and source of microglia autofluorescence, we did confocal imaging and spectral analysis of microglia autofluorescence on fixed brain tissues, revealing that microglia autofluorescence emitted from cytoplasmic granules and displayed a multi-peak emission spectrum. We recommended removing autofluorescence by lipofuscin removing agents when staining intracellular proteins in microglia with the immunofluorescence techniques. On live brain slices, autofluorescent granules reduced the amplitudes of calcium signals in microglial somata derived from GCaMP6s fluorescence and thus needed to be excluded when selecting regions of interest (ROI). CONCLUSIONS In conclusion, autofluorescence is a critical factor to consider when designing experiments and interpreting results based on fluorescence-based techniques to study microglia.
Collapse
Affiliation(s)
- Haozhe Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.,Translational Medicine Research Center, People's Hospital Of Zhengzhou, Zhengzhou, Henan, China
| | - Chen Tan
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoyue Shi
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ji Xu
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China. .,Institute of Neuroscience, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
20
|
Rieder P, Gobbo D, Stopper G, Welle A, Damo E, Kirchhoff F, Scheller A. Astrocytes and Microglia Exhibit Cell-Specific Ca2+ Signaling Dynamics in the Murine Spinal Cord. Front Mol Neurosci 2022; 15:840948. [PMID: 35431801 PMCID: PMC9006623 DOI: 10.3389/fnmol.2022.840948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/22/2022] [Indexed: 12/31/2022] Open
Abstract
The spinal cord is the main pathway connecting brain and peripheral nervous system. Its functionality relies on the orchestrated activity of both neurons and glial cells. To date, most advancement in understanding the spinal cord inner mechanisms has been made either by in vivo exposure of its dorsal surface through laminectomy or by acute ex vivo slice preparation, likely affecting spinal cord physiology in virtue of the necessary extensive manipulation of the spinal cord tissue. This is especially true of cells immediately responding to alterations of the surrounding environment, such as microglia and astrocytes, reacting within seconds or minutes and for up to several days after the original insult. Ca2+ signaling is considered one of the most immediate, versatile, and yet elusive cellular responses of glia. Here, we induced the cell-specific expression of the genetically encoded Ca2+ indicator GCaMP3 to evaluate spontaneous intracellular Ca2+ signaling in astrocytes and microglia. Ca2+ signals were then characterized in acute ex vivo (both gray and white matter) as well as in chronic in vivo (white matter) preparations using MSparkles, a MATLAB-based software for automatic detection and analysis of fluorescence events. As a result, we were able to segregate distinct astroglial and microglial Ca2+ signaling patterns along with method-specific Ca2+ signaling alterations, which must be taken into consideration in the reliable evaluation of any result obtained in physiological as well as pathological conditions. Our study revealed a high degree of Ca2+ signaling diversity in glial cells of the murine spinal cord, thus adding to the current knowledge of the astonishing glial heterogeneity and cell-specific Ca2+ dynamics in non-neuronal networks.
Collapse
Affiliation(s)
- Phillip Rieder
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Gebhard Stopper
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Anna Welle
- Department of Genetics and Epigenetics, University of Saarland, Saarbrücken, Germany
| | - Elisa Damo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Anja Scheller
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
- *Correspondence: Anja Scheller,
| |
Collapse
|
21
|
Bolton JL, Short AK, Othy S, Kooiker CL, Shao M, Gunn BG, Beck J, Bai X, Law SM, Savage JC, Lambert JJ, Belelli D, Tremblay MÈ, Cahalan MD, Baram TZ. Early stress-induced impaired microglial pruning of excitatory synapses on immature CRH-expressing neurons provokes aberrant adult stress responses. Cell Rep 2022; 38:110600. [PMID: 35354026 PMCID: PMC9014810 DOI: 10.1016/j.celrep.2022.110600] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Several mental illnesses, characterized by aberrant stress reactivity, often arise after early-life adversity (ELA). However, it is unclear how ELA affects stress-related brain circuit maturation, provoking these enduring vulnerabilities. We find that ELA increases functional excitatory synapses onto stress-sensitive hypothalamic corticotropin-releasing hormone (CRH)-expressing neurons, resulting from disrupted developmental synapse pruning by adjacent microglia. Microglial process dynamics and synaptic element engulfment were attenuated in ELA mice, associated with deficient signaling of the microglial phagocytic receptor MerTK. Accordingly, selective chronic chemogenetic activation of ELA microglia increased microglial process dynamics and reduced excitatory synapse density to control levels. Notably, selective early-life activation of ELA microglia normalized adult acute and chronic stress responses, including stress-induced hormone secretion and behavioral threat responses, as well as chronic adrenal hypertrophy of ELA mice. Thus, microglial actions during development are powerful contributors to mechanisms by which ELA sculpts the connectivity of stress-regulating neurons, promoting vulnerability to stress and stress-related mental illnesses.
Collapse
Affiliation(s)
- Jessica L Bolton
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA.
| | - Annabel K Short
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Cassandra L Kooiker
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Manlin Shao
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Benjamin G Gunn
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA; Division of Neuroscience, Medical Research Institute, Dundee University, Ninewells Hospital and Medical School, Dundee, UK
| | - Jaclyn Beck
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Xinglong Bai
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Stephanie M Law
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Julie C Savage
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada; Axe Neurosciences, Centre de recherche du CHU de Québec, Québec City, QC, Canada
| | - Jeremy J Lambert
- Division of Neuroscience, Medical Research Institute, Dundee University, Ninewells Hospital and Medical School, Dundee, UK
| | - Delia Belelli
- Division of Neuroscience, Medical Research Institute, Dundee University, Ninewells Hospital and Medical School, Dundee, UK
| | - Marie-Ève Tremblay
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada; Axe Neurosciences, Centre de recherche du CHU de Québec, Québec City, QC, Canada
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
22
|
Microglia Modulate Cortical Spreading Depolarizations After Ischemic Stroke: A Narrative Review. Neurocrit Care 2022; 37:133-138. [PMID: 35288861 PMCID: PMC9259539 DOI: 10.1007/s12028-022-01469-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/08/2022] [Indexed: 01/06/2023]
Abstract
Cortical spreading depolarizations (CSDs) are characterized by waves of diminished electroencephalography activity that propagate across the cortex with subsequent loss of ionic homeostasis. CSDs have been found in many pathological conditions, including migraine, traumatic brain injury, and ischemic stroke. Because of CSD-associated ionic and metabolic disturbances at the peri-infarct area after ischemic stroke, it is thought that CSDs exacerbate tissue infarction and worsen clinical outcomes. Microglia, the main innate immune cells in the brain, are among the first responders to brain tissue damage. Recent studies demonstrated that microglia play a critical role in CSD initiation and propagation. In this article, we discuss the significance of CSD in the setting of ischemic stroke and how microglia may modulate peri-infarct CSDs, also known as iso-electric depolarizations. Finally, we discuss the significance of microglial Ca2+ and how it might be used as a potential therapeutic target for patients with ischemic stroke.
Collapse
|
23
|
Logiacco F, Xia P, Georgiev SV, Franconi C, Chang YJ, Ugursu B, Sporbert A, Kühn R, Kettenmann H, Semtner M. Microglia sense neuronal activity via GABA in the early postnatal hippocampus. Cell Rep 2021; 37:110128. [PMID: 34965412 DOI: 10.1016/j.celrep.2021.110128] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/14/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023] Open
Abstract
Microglia, the resident macrophages in the central nervous system, express receptors for classical neurotransmitters, such as γ-aminobutyric acid (GABA) and glutamate, suggesting that they sense synaptic activity. To detect microglial Ca2+ responses to neuronal activity, we generate transgenic mouse lines expressing the fluorescent Ca2+ indicator GCaMP6m, specifically in microglia and demonstrate that electrical stimulation of the Schaffer collateral pathway results in microglial Ca2+ responses in early postnatal but not adult hippocampus. Preceding the microglial responses, we also observe similar Ca2+ responses in astrocytes, and both are sensitive to tetrodotoxin. Blocking astrocytic glutamate uptake or GABA transport abolishes stimulation-induced microglial responses as well as antagonizing the microglial GABAB receptor. Our data, therefore, suggest that the neuronal activity-induced glutamate uptake and the release of GABA by astrocytes trigger the activation of GABAB receptors in microglia. This neuron, astrocyte, and microglia communication pathway might modulate microglial activity in developing neuronal networks.
Collapse
Affiliation(s)
- Francesca Logiacco
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 12169 Berlin, Germany
| | - Pengfei Xia
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Svilen Veselinov Georgiev
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Celeste Franconi
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Yi-Jen Chang
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Bilge Ugursu
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Ralf Kühn
- Transgenic Core Facility, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.
| |
Collapse
|
24
|
Tworig JM, Coate C, Feller MB. Excitatory neurotransmission activates compartmentalized calcium transients in Müller glia without affecting lateral process motility. eLife 2021; 10:73202. [PMID: 34913435 PMCID: PMC8806189 DOI: 10.7554/elife.73202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Neural activity has been implicated in the motility and outgrowth of glial cell processes throughout the central nervous system. Here, we explore this phenomenon in Müller glia, which are specialized radial astroglia that are the predominant glial type of the vertebrate retina. Müller glia extend fine filopodia-like processes into retinal synaptic layers, in similar fashion to brain astrocytes and radial glia that exhibit perisynaptic processes. Using two-photon volumetric imaging, we found that during the second postnatal week, Müller glial processes were highly dynamic, with rapid extensions and retractions that were mediated by cytoskeletal rearrangements. During this same stage of development, retinal waves led to increases in cytosolic calcium within Müller glial lateral processes and stalks. These regions comprised distinct calcium compartments, distinguished by variable participation in waves, timing, and sensitivity to an M1 muscarinic acetylcholine receptor antagonist. However, we found that motility of lateral processes was unaffected by the presence of pharmacological agents that enhanced or blocked wave-associated calcium transients. Finally, we found that mice lacking normal cholinergic waves in the first postnatal week also exhibited normal Müller glial process morphology. Hence, outgrowth of Müller glial lateral processes into synaptic layers is determined by factors that are independent of neuronal activity. When it comes to studying the nervous system, neurons often steal the limelight; yet, they can only work properly thanks to an ensemble cast of cell types whose roles are only just emerging. For example, ‘glial cells’ – their name derives from the Greek word for glue – were once thought to play only a passive, supporting function in nervous tissues. Now, growing evidence shows that they are, in fact, integrated into neural circuits: their activity is influenced by neurons, and, in turn, they help neurons to function properly. The role of glial cells is becoming clear in the retina, the thin, light-sensitive layer that lines the back of the eye and relays visual information to the brain. There, beautifully intricate Müller glial cells display fine protrusions (or ‘processes') that intermingle with synapses, the busy space between neurons where chemical messengers are exchanged. These messengers can act on Müller cells, triggering cascades of molecular events that may influence the structure and function of glia. This is of particular interest during development: as Müller cells mature, they are exposed to chemicals released by more fully formed retinal neurons. Tworig et al. explored how neuronal messengers can influence the way Müller cells grow their processes. To do so, they tracked mouse retinal glial cells ‘live’ during development, showing that they were growing fine, highly dynamic processes in a region rich in synapses just as neurons and glia increased their communication. However, using drugs to disrupt this messaging for a short period did not seem to impact how the processes grew. Extending the blockade over a longer timeframe also did not change the way Müller cells developed, with the cells still acquiring their characteristic elaborate process networks. Taken together, these results suggest that the structural maturation of Müller glial cells is not impacted by neuronal signaling, giving a more refined understanding of how glia form in the retina and potentially in the brain.
Collapse
Affiliation(s)
- Joshua M Tworig
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Chandler Coate
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Marla B Feller
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
25
|
Altered synaptic connectivity and brain function in mice lacking microglial adapter protein Iba1. Proc Natl Acad Sci U S A 2021; 118:2115539118. [PMID: 34764226 DOI: 10.1073/pnas.2115539118] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 12/24/2022] Open
Abstract
Growing evidence indicates that microglia impact brain function by regulating synaptic pruning and formation as well as synaptic transmission and plasticity. Iba1 (ionized Ca+2-binding adapter protein 1), encoded by the Allograft inflammatory factor 1 (Aif1) gene, is an actin-interacting protein in microglia. Although Iba1 has long been used as a cellular marker for microglia, its functional role remains unknown. Here, we used global, Iba1-deficient (Aif1 -/-) mice to characterize microglial activity, synaptic function, and behavior. Microglial imaging in acute hippocampal slices and fixed tissues from juvenile mice revealed that Aif1 -/- microglia display reductions in ATP-induced motility and ramification, respectively. Biochemical assays further demonstrated that Aif1 -/- brain tissues exhibit an altered expression of microglial-enriched proteins associated with synaptic pruning. Consistent with these changes, juvenile Aif1 -/- mice displayed deficits in the excitatory synapse number and synaptic drive assessed by neuronal labeling and whole-cell patch-clamp recording in acute hippocampal slices. Unexpectedly, microglial synaptic engulfment capacity was diminished in juvenile Aif1 -/- mice. During early postnatal development, when synapse formation is a predominant event in the hippocampus, the excitatory synapse number was still reduced in Aif1 -/- mice. Together, these findings support an overall role of Iba1 in excitatory synaptic growth in juvenile mice. Lastly, postnatal synaptic deficits persisted in adulthood and correlated with significant behavioral changes in adult Aif1 -/- mice, which exhibited impairments in object recognition memory and social interaction. These results suggest that Iba1 critically contributes to microglial activity underlying essential neuroglia developmental processes that may deeply influence behavior.
Collapse
|
26
|
Effects of Metformin on Spontaneous Ca 2+ Signals in Cultured Microglia Cells under Normoxic and Hypoxic Conditions. Int J Mol Sci 2021; 22:ijms22179493. [PMID: 34502402 PMCID: PMC8430509 DOI: 10.3390/ijms22179493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/20/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022] Open
Abstract
Microglial functioning depends on Ca2+ signaling. By using Ca2+ sensitive fluorescence dye, we studied how inhibition of mitochondrial respiration changed spontaneous Ca2+ signals in soma of microglial cells from 5-7-day-old rats grown under normoxic and mild-hypoxic conditions. In microglia under normoxic conditions, metformin or rotenone elevated the rate and the amplitude of Ca2+ signals 10-15 min after drug application. Addition of cyclosporin A, a blocker of mitochondrial permeability transition pore (mPTP), antioxidant trolox, or inositol 1,4,5-trisphosphate receptor (IP3R) blocker caffeine in the presence of rotenone reduced the elevated rate and the amplitude of the signals implying sensitivity to reactive oxygen species (ROS), and involvement of mitochondrial mPTP together with IP3R. Microglial cells exposed to mild hypoxic conditions for 24 h showed elevated rate and increased amplitude of Ca2+ signals. Application of metformin or rotenone but not phenformin before mild hypoxia reduced this elevated rate. Thus, metformin and rotenone had the opposing fast action in normoxia after 10-15 min and the slow action during 24 h mild-hypoxia implying activation of different signaling pathways. The slow action of metformin through inhibition of complex I could stabilize Ca2+ homeostasis after mild hypoxia and could be important for reduction of ischemia-induced microglial activation.
Collapse
|
27
|
Odoj K, Brawek B, Asavapanumas N, Mojtahedi N, Heneka MT, Garaschuk O. In vivo mechanisms of cortical network dysfunction induced by systemic inflammation. Brain Behav Immun 2021; 96:113-126. [PMID: 34052361 DOI: 10.1016/j.bbi.2021.05.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/22/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022] Open
Abstract
Peripheral inflammation is known to impact brain function, resulting in lethargy, loss of appetite and impaired cognitive abilities. However, the channels for information transfer from the periphery to the brain, the corresponding signaling molecules and the inflammation-induced interaction between microglia and neurons remain obscure. Here, we used longitudinal in vivo two-photon Ca2+ imaging to monitor neuronal activity in the mouse cortex throughout the early (initiation) and late (resolution) phases of peripheral inflammation. Single peripheral lipopolysaccharide injection induced a substantial but transient increase in ongoing neuronal activity, restricted to the initiation phase, whereas the impairment of visual processing was selectively observed during the resolution phase of systemic inflammation. In the frontal/motor cortex, the initiation phase-specific cortical hyperactivity was seen in the deep (layer 5) and superficial (layer 2/3) pyramidal neurons but not in the axons coming from the somatosensory cortex, and was accompanied by reduced activity of layer 2/3 cortical interneurons. Moreover, the hyperactivity was preserved after depletion of microglia and in NLRP3-/- mice but absent in TNF-α-/- mice. Together, these data identify microglia-independent and TNF-α-mediated reduction of cortical inhibition as a likely cause of the initiation phase-specific cortical hyperactivity and reveal the resolution phase-specific impairment of sensory processing, presumably caused by activated microglia.
Collapse
Affiliation(s)
- Karin Odoj
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Bianca Brawek
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nithi Asavapanumas
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nima Mojtahedi
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Olga Garaschuk
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University Tübingen, Tübingen, Germany.
| |
Collapse
|
28
|
Melo PN, Souza da Silveira M, Mendes Pinto I, Relvas JB. Morphofunctional programming of microglia requires distinct roles of type II myosins. Glia 2021; 69:2717-2738. [PMID: 34329508 DOI: 10.1002/glia.24067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 11/05/2022]
Abstract
The ramified morphology of microglia and the dynamics of their membrane protrusions are essential for their functions in central nervous system development, homeostasis, and disease. Although their ability to change and control shape critically depends on the actin and actomyosin cytoskeleton, the underlying regulatory mechanisms remain largely unknown. In this study, we systematically analyzed the actomyosin cytoskeleton and regulators downstream of the small GTPase RhoA in the control of microglia shape and function. Our results reveal that (i) Myh9 controls cortical tension levels and affects microglia protrusion formation, (ii) cofilin-mediated maintenance of actin turnover regulates microglia protrusion extension, and (iii) Myh10 influences microglia inflammatory activation. Overall we uncover molecular pathways that regulate microglia morphology and identify type-II myosins as important regulators of microglia biology with differential roles in the control of cell shape (Myh9) and functions (Myh10).
Collapse
Affiliation(s)
- Pedro Neves Melo
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Graduate Programme in Areas of Basic and Applied Biology (GABBA), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Mariana Souza da Silveira
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Instituto de Biofísica Carlos Chagas Filho (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Inês Mendes Pinto
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Life Sciences, International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - João Bettencourt Relvas
- Instituto de Investigação e Inovação em Saúde (I3S), Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal.,Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
29
|
Redmon SN, Yarishkin O, Lakk M, Jo A, Mustafic E, Tvrdik P, Križaj D. TRPV4 channels mediate the mechanoresponse in retinal microglia. Glia 2021; 69:1563-1582. [PMID: 33624376 PMCID: PMC8989051 DOI: 10.1002/glia.23979] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022]
Abstract
The physiological and neurological correlates of plummeting brain osmolality during edema, traumatic CNS injury, and severe ischemia are compounded by neuroinflammation. Using multiple approaches, we investigated how retinal microglia respond to challenges mediated by increases in strain, osmotic gradients, and agonists of the stretch-activated cation channel TRPV4. Dissociated and intact microglia were TRPV4-immunoreactive and responded to the selective agonist GSK1016790A and substrate stretch with altered motility and elevations in intracellular calcium ([Ca2+ ]i ). Agonist- and hypotonicity-induced swelling was associated with a nonselective outwardly rectifying cation current, increased [Ca2+ ]i , and retraction of higher-order processes. The antagonist HC067047 reduced the extent of hypotonicity-induced microglial swelling and inhibited the suppressive effects of GSK1016790A and hypotonicity on microglial branching. Microglial TRPV4 signaling required intermediary activation of phospholipase A2 (PLA2), cytochrome P450, and epoxyeicosatrienoic acid production (EETs). The expression pattern of vanilloid thermoTrp genes in retinal microglia was markedly different from retinal neurons, astrocytes, and cortical microglia. These results suggest that TRPV4 represents a primary retinal microglial sensor of osmochallenges under physiological and pathological conditions. Its activation, associated with PLA2, modulates calcium signaling and cell architecture. TRPV4 inhibition might be a useful strategy to suppress microglial overactivation in the swollen and edematous CNS.
Collapse
Affiliation(s)
- Sarah N. Redmon
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Oleg Yarishkin
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Monika Lakk
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Andrew Jo
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Edin Mustafic
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
| | - Peter Tvrdik
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville VA 22908
| | - David Križaj
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, Salt Lake City, UT 84132
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT 84132
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84132
- Department of Neurobiology & Anatomy, University of Utah, Salt Lake City, UT 84132
| |
Collapse
|
30
|
Andoh M, Koyama R. Assessing Microglial Dynamics by Live Imaging. Front Immunol 2021; 12:617564. [PMID: 33763064 PMCID: PMC7982483 DOI: 10.3389/fimmu.2021.617564] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Microglia are highly dynamic in the brain in terms of their ability to migrate, proliferate, and phagocytose over the course of an individual's life. Real-time imaging is a useful tool to examine how microglial behavior is regulated and how it affects the surrounding environment. However, microglia are sensitive to environmental stimuli, so they possibly change their state during live imaging in vivo, mainly due to surgical damage, and in vitro due to various effects associated with culture conditions. Therefore, it is difficult to perform live imaging without compromising the properties of the microglia under physiological conditions. To overcome this barrier, various experimental conditions have been developed; recently, it has become possible to perform live imaging of so-called surveillant microglia in vivo, ex vivo, and in vitro, although there are various limitations. Now, we can choose in vivo, ex vivo, or in vitro live imaging systems according to the research objective. In this review, we discuss the advantages and disadvantages of each experimental system and outline the physiological significance and molecular mechanisms of microglial behavior that have been elucidated by live imaging.
Collapse
Affiliation(s)
- Megumi Andoh
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
31
|
Changing Functional Signatures of Microglia along the Axis of Brain Aging. Int J Mol Sci 2021; 22:ijms22031091. [PMID: 33499206 PMCID: PMC7865559 DOI: 10.3390/ijms22031091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/19/2022] Open
Abstract
Microglia, the innate immune cells of the brain, are commonly perceived as resident macrophages of the central nervous system (CNS). This definition, however, requires further specification, as under healthy homeostatic conditions, neither morphological nor functional properties of microglia mirror those of classical macrophages. Indeed, microglia adapt exceptionally well to their microenvironment, becoming a legitimate member of the cellular brain architecture. The ramified or surveillant microglia in the young adult brain are characterized by specific morphology (small cell body and long, thin motile processes) and physiology (a unique pattern of Ca2+ signaling, responsiveness to various neurotransmitters and hormones, in addition to classic “immune” stimuli). Their numerous physiological functions far exceed and complement their immune capabilities. As the brain ages, the respective changes in the microglial microenvironment impact the functional properties of microglia, triggering further rounds of adaptation. In this review, we discuss the recent data showing how functional properties of microglia adapt to age-related changes in brain parenchyma in a sex-specific manner, with a specific focus on early changes occurring at middle age as well as some strategies counteracting the aging of microglia.
Collapse
|
32
|
Laprell L, Schulze C, Brehme ML, Oertner TG. The role of microglia membrane potential in chemotaxis. J Neuroinflammation 2021; 18:21. [PMID: 33423699 PMCID: PMC7798195 DOI: 10.1186/s12974-020-02048-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/09/2020] [Indexed: 11/10/2022] Open
Abstract
Microglia react to danger signals by rapid and targeted extension of cellular processes towards the source of the signal. This positive chemotactic response is accompanied by a hyperpolarization of the microglia membrane. Here, we show that optogenetic depolarization of microglia has little effect on baseline motility, but significantly slows down the chemotactic response. Reducing the extracellular Ca2+ concentration mimics the effect of optogenetic depolarization. As the membrane potential sets the driving force for Ca2+ entry, hyperpolarization is an integral part of rapid stimulus-response coupling in microglia. Compared to typical excitable cells such as neurons, the sign of the activating response is inverted in microglia, leading to inhibition by depolarizing channelrhodopsins.
Collapse
Affiliation(s)
- Laura Laprell
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany.
| | - Christian Schulze
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Marie-Luise Brehme
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Thomas G Oertner
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany.
| |
Collapse
|
33
|
Hopp SC. Targeting microglia L-type voltage-dependent calcium channels for the treatment of central nervous system disorders. J Neurosci Res 2021; 99:141-162. [PMID: 31997405 PMCID: PMC9394523 DOI: 10.1002/jnr.24585] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022]
Abstract
Calcium (Ca2+ ) is a ubiquitous mediator of a multitude of cellular functions in the central nervous system (CNS). Intracellular Ca2+ is tightly regulated by cells, including entry via plasma membrane Ca2+ permeable channels. Of specific interest for this review are L-type voltage-dependent Ca2+ channels (L-VDCCs), due to their pleiotropic role in several CNS disorders. Currently, there are numerous approved drugs that target L-VDCCs, including dihydropyridines. These drugs are safe and effective for the treatment of humans with cardiovascular disease and may also confer neuroprotection. Here, we review the potential of L-VDCCs as a target for the treatment of CNS disorders with a focus on microglia L-VDCCs. Microglia, the resident immune cells of the brain, have attracted recent attention for their emerging inflammatory role in several CNS diseases. Intracellular Ca2+ regulates microglia transition from a resting quiescent state to an "activated" immune-effector state and is thus a valuable target for manipulation of microglia phenotype. We will review the literature on L-VDCC expression and function in the CNS and on microglia in vitro and in vivo and explore the therapeutic landscape of L-VDCC-targeting agents at present and future challenges in the context of Alzheimer's disease, Parkinson's disease, Huntington's disease, neuropsychiatric diseases, and other CNS disorders.
Collapse
Affiliation(s)
- Sarah C. Hopp
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| |
Collapse
|
34
|
Umpierre AD, Wu LJ. How microglia sense and regulate neuronal activity. Glia 2020; 69:1637-1653. [PMID: 33369790 DOI: 10.1002/glia.23961] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022]
Abstract
Microglia are innate immune cells of the central nervous system that sense extracellular cues. Brain injuries, inflammation, and pathology evoke dynamic structural responses in microglia, altering their morphology and motility. The dynamic motility of microglia is hypothesized to be a critical first step in sensing local alterations and engaging in pattern-specific responses. Alongside their pathological responses, microglia also sense and regulate neuronal activity. In this review, we consider the extracellular molecules, receptors, and mechanisms that allow microglia to sense neuronal activity changes under both hypoactivity and hyperactivity. We also highlight emerging in vivo evidence that microglia regulate neuronal activity, ranging from physiological to pathophysiological conditions. In addition, we discuss the emerging role of calcium signaling in microglial responses to the extracellular environment. The dynamic function of microglia in monitoring and influencing neuronal activity may be critical for brain homeostasis and circuit modification in health and disease.
Collapse
Affiliation(s)
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA.,Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA.,Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
35
|
Liang Y, Walczak P. Long term intravital single cell tracking under multiphoton microscopy. J Neurosci Methods 2020; 349:109042. [PMID: 33340557 DOI: 10.1016/j.jneumeth.2020.109042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Visualizing and tracking cells over time in a living organism has been a much-coveted dream before the invention of intravital microscopy. The opaque nature of tissue was a major hurdle that was remedied by the multiphoton microscopy. With the advancement of optical imaging and fluorescent labeling tools, intravital high resolution imaging has become increasingly accessible over the past few years. Long-term intravital tracking of single cells (LIST) under multiphoton microscopy provides a unique opportunity to gain insight into the longitudinal changes in the morphology, migration, or function of cells or subcellular structures. It is particularly suitable for studying slow-evolving cellular and molecular events during normal development or disease progression, without losing the opportunity of catching fast events such as calcium signals. Here, we review the application of LIST under 2-photon microscopy in various fields of neurobiology and discuss challenges and new directions in labeling and imaging methods for LIST. Overall, this review provides an overview of current applications of LIST in mammals, which is an emerging field that will contribute to a better understanding of essential molecular and cellular events in health and disease.
Collapse
Affiliation(s)
- Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
Shipley FB, Dani N, Xu H, Deister C, Cui J, Head JP, Sadegh C, Fame RM, Shannon ML, Flores VI, Kishkovich T, Jang E, Klein EM, Goldey GJ, He K, Zhang Y, Holtzman MJ, Kirchhausen T, Wyart C, Moore CI, Andermann ML, Lehtinen MK. Tracking Calcium Dynamics and Immune Surveillance at the Choroid Plexus Blood-Cerebrospinal Fluid Interface. Neuron 2020; 108:623-639.e10. [PMID: 32961128 PMCID: PMC7847245 DOI: 10.1016/j.neuron.2020.08.024] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 05/18/2020] [Accepted: 08/25/2020] [Indexed: 12/26/2022]
Abstract
The choroid plexus (ChP) epithelium is a source of secreted signaling factors in cerebrospinal fluid (CSF) and a key barrier between blood and brain. Here, we develop imaging tools to interrogate these functions in adult lateral ventricle ChP in whole-mount explants and in awake mice. By imaging epithelial cells in intact ChP explants, we observed calcium activity and secretory events that increased in frequency following delivery of serotonergic agonists. Using chronic two-photon imaging in awake mice, we observed spontaneous subcellular calcium events as well as strong agonist-evoked calcium activation and cytoplasmic secretion into CSF. Three-dimensional imaging of motility and mobility of multiple types of ChP immune cells at baseline and following immune challenge or focal injury revealed a range of surveillance and defensive behaviors. Together, these tools should help illuminate the diverse functions of this understudied body-brain interface.
Collapse
Affiliation(s)
- Frederick B Shipley
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Neil Dani
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Huixin Xu
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Christopher Deister
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Jin Cui
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Joshua P Head
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Cameron Sadegh
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Morgan L Shannon
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Vanessa I Flores
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Thomas Kishkovich
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Emily Jang
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Eric M Klein
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Glenn J Goldey
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Kangmin He
- Department of Cell Biology and Department of Pediatrics, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Tomas Kirchhausen
- Department of Cell Biology and Department of Pediatrics, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Claire Wyart
- Institut du Cerveau et de la Moelle Épinière (ICM), Sorbonne Université, Inserm U1127, CNRS UMR 7225, 75013 Paris, France
| | - Christopher I Moore
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Mark L Andermann
- Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA.
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
37
|
Liu L, Kearns KN, Eli I, Sharifi KA, Soldozy S, Carlson EW, Scott KW, Sluzewski MF, Acton ST, Stauderman KA, Kalani MYS, Park M, Tvrdik P. Microglial Calcium Waves During the Hyperacute Phase of Ischemic Stroke. Stroke 2020; 52:274-283. [PMID: 33161850 DOI: 10.1161/strokeaha.120.032766] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND PURPOSE Ischemic injury triggers multiple pathological responses in the brain tissue, including spreading depolarizations across the cerebral cortex (cortical spreading depolarizations [CSD]). Microglia have been recently shown to play a significant role in the propagation of CSD. However, the intracellular responses of myeloid cells during ischemic stroke have not been investigated. METHODS We have studied intracellular calcium activity in cortical microglia in the stroke model of the middle cerebral artery occlusion, using the murine Polr2a-based and Cre-dependent GCaMP5 and tdTomato reporter (PC::G5-tdT). High-speed 2-photon microscopy through cranial windows was employed to record signals from genetically encoded indicators of calcium. Inflammatory stimuli and pharmacological inhibition were used to modulate microglial calcium responses in the somatosensory cortex. RESULTS In vivo imaging revealed periodical calcium activity in microglia during the hyperacute phase of ischemic stroke. This activity was more frequent during the first 6 hours after occlusion, but the amplitudes of calcium transients became larger at later time points. Consistent with CSD nature of these events, we reproducibly triggered comparable calcium transients with microinjections of potassium chloride (KCl) into adjacent cortical areas. Furthermore, lipopolysaccharide-induced peripheral inflammation, mimicking sterile inflammation during ischemic stroke, produced significantly greater microglial calcium transients during CSD. Finally, in vivo pharmacological analysis with CRAC (calcium release-activated channel) inhibitor CM-EX-137 demonstrated that CSD-associated microglial calcium transients after KCl microinjections are mediated at least in part by the CRAC mechanism. CONCLUSIONS Our findings demonstrate that microglia participate in ischemic brain injury via previously undetected mechanisms, which may provide new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Lei Liu
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - Kathryn N Kearns
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - Ilyas Eli
- Department of Neurosurgery (I.E., E.W.C.), University of Utah School of Medicine, Salt Lake City
| | - Khadijeh A Sharifi
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
- Department of Neuroscience (K.A. Sharifi, M.Y.S.K., P.T.), University of Virginia Health System, Charlottesville
| | - Sauson Soldozy
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - Elizabeth W Carlson
- Department of Neurosurgery (I.E., E.W.C.), University of Utah School of Medicine, Salt Lake City
| | - Kyle W Scott
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - M Filip Sluzewski
- Department of Electrical and Computer Engineering (M.F.S., S.T.A.), University of Virginia Health System, Charlottesville
| | - Scott T Acton
- Department of Electrical and Computer Engineering (M.F.S., S.T.A.), University of Virginia Health System, Charlottesville
| | | | - M Yashar S Kalani
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
- Department of Neuroscience (K.A. Sharifi, M.Y.S.K., P.T.), University of Virginia Health System, Charlottesville
| | - Min Park
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
| | - Petr Tvrdik
- Department of Neurological Surgery (L.L., K.N.K., K.A. Sharifi, S.S., K.W.S., M.Y.S.K., M.P., P.T.), University of Virginia Health System, Charlottesville
- Department of Neuroscience (K.A. Sharifi, M.Y.S.K., P.T.), University of Virginia Health System, Charlottesville
| |
Collapse
|
38
|
Phosphoinositide-3-Kinase γ Is Not a Predominant Regulator of ATP-Dependent Directed Microglial Process Motility or Experience-Dependent Ocular Dominance Plasticity. eNeuro 2020; 7:ENEURO.0311-20.2020. [PMID: 33067365 PMCID: PMC7769883 DOI: 10.1523/eneuro.0311-20.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/16/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022] Open
Abstract
Microglia are dynamic cells whose extensive interactions with neurons and glia during development allow them to regulate neuronal development and function. The microglial P2Y12 receptor is crucial for microglial responsiveness to extracellular ATP and mediates numerous microglial functions, including ATP-dependent directional motility, microglia-neuron interactions, and experience-dependent synaptic plasticity. However, little is known about the downstream signaling effectors that mediate these diverse actions of P2Y12. Phosphoinositide-3-kinase γ (PI3Kγ), a lipid kinase activated downstream of Gi-protein-coupled receptors such as P2Y12, could translate localized extracellular ATP signals into directed microglial action and serve as a broad effector of P2Y12-dependent signaling. Here, we used pharmacological and genetic methods to manipulate P2Y12 and PI3Kγ signaling to determine whether inhibiting PI3Kγ phenocopied the loss of P2Y12 signaling in mouse microglia. While pan-inhibition of all PI3K activity substantially affected P2Y12-dependent microglial responses, our results suggest that PI3Kγ specifically is only a minor part of the P2Y12 signaling pathway. PI3Kγ was not required to maintain homeostatic microglial morphology or their dynamic surveillance in vivo Further, PI3Kγ was not strictly required for P2Y12-dependent microglial responses ex vivo or in vivo, although we did observe subtle deficits in the recruitment of microglial process toward sources of ATP. Finally, PI3Kγ was not required for ocular dominance plasticity, a P2Y12-dependent form of experience-dependent synaptic plasticity that occurs in the developing visual cortex. Overall, our results demonstrate that PI3Kγ is not the major mediator of P2Y12 function in microglia, but may have a role in amplifying or fine-tuning the chemotactic response.
Collapse
|
39
|
Hughes AN, Appel B. Microglia phagocytose myelin sheaths to modify developmental myelination. Nat Neurosci 2020; 23:1055-1066. [PMID: 32632287 PMCID: PMC7483351 DOI: 10.1038/s41593-020-0654-2] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 05/13/2020] [Indexed: 12/15/2022]
Abstract
During development, oligodendrocytes contact and wrap neuronal axons with myelin. Similarly to neurons and synapses, excess myelin sheaths are produced and selectively eliminated, but how elimination occurs is unknown. Microglia, the resident immune cells of the central nervous system, engulf surplus neurons and synapses. To determine whether microglia also prune myelin sheaths, we used zebrafish to visualize and manipulate interactions between microglia, oligodendrocytes, and neurons during development. We found that microglia closely associate with oligodendrocytes and specifically phagocytose myelin sheaths. By using a combination of optical, genetic, chemogenetic, and behavioral approaches, we reveal that neuronal activity bidirectionally balances microglial association with neuronal cell bodies and myelin phagocytosis in the optic tectum. Furthermore, multiple strategies to deplete microglia resulted in oligodendrocytes maintaining excessive and ectopic myelin. Our work reveals a neuronal activity-regulated role for microglia in modifying developmental myelin targeting by oligodendrocytes.
Collapse
Affiliation(s)
| | - Bruce Appel
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, Aurora, CO, USA.
| |
Collapse
|
40
|
Bernier LP, Bohlen CJ, York EM, Choi HB, Kamyabi A, Dissing-Olesen L, Hefendehl JK, Collins HY, Stevens B, Barres BA, MacVicar BA. Nanoscale Surveillance of the Brain by Microglia via cAMP-Regulated Filopodia. Cell Rep 2020; 27:2895-2908.e4. [PMID: 31167136 DOI: 10.1016/j.celrep.2019.05.010] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/14/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023] Open
Abstract
Microglia, the brain's immune cells, maintain homeostasis and sense pathological changes by continuously surveying the parenchyma with highly motile large processes. Here, we demonstrate that microglia also use thin actin-dependent filopodia that allow fast nanoscale sensing within discrete regions. Filopodia are distinct from large processes by their size, speed, and regulation mechanism. Increasing cyclic AMP (cAMP) by activating norepinephrine Gs-coupled receptors, applying nitric oxide, or inhibiting phosphodiesterases rapidly increases filopodia but collapses large processes. Alternatively, Gi-coupled P2Y12 receptor activation collapses filopodia but triggers large processes extension with bulbous tips. Similar control of cytoskeletal dynamics and microglial morphology by cAMP is observed in ramified primary microglia, suggesting that filopodia are intrinsically generated sensing structures. Therefore, nanoscale surveillance of brain parenchyma by microglia requires localized cAMP increases that drive filopodia formation. Shifting intracellular cAMP levels controls the polarity of microglial responses to changes in brain homeostasis and alters the scale of immunosurveillance.
Collapse
Affiliation(s)
- Louis-Philippe Bernier
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada.
| | - Christopher J Bohlen
- Stanford University School of Medicine, Department of Neurobiology, Stanford, CA 94305, USA
| | - Elisa M York
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Hyun B Choi
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Alireza Kamyabi
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Lasse Dissing-Olesen
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School Boston, MA 02115, USA
| | - Jasmin K Hefendehl
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Hannah Y Collins
- Stanford University School of Medicine, Department of Neurobiology, Stanford, CA 94305, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School Boston, MA 02115, USA
| | - Ben A Barres
- Stanford University School of Medicine, Department of Neurobiology, Stanford, CA 94305, USA
| | - Brian A MacVicar
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
41
|
Umpierre AD, Bystrom LL, Ying Y, Liu YU, Worrell G, Wu LJ. Microglial calcium signaling is attuned to neuronal activity in awake mice. eLife 2020; 9:56502. [PMID: 32716294 PMCID: PMC7402678 DOI: 10.7554/elife.56502] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
Microglial calcium signaling underlies a number of key physiological and pathological processes in situ, but has not been studied in vivo in awake mice. Using multiple GCaMP6 variants targeted to microglia, we assessed how microglial calcium signaling responds to alterations in neuronal activity across a wide range. We find that only a small subset of microglial somata and processes exhibited spontaneous calcium transients in a chronic window preparation. However, hyperactive shifts in neuronal activity (kainate status epilepticus and CaMKIIa Gq DREADD activation) triggered increased microglial process calcium signaling, often concomitant with process extension. Additionally, hypoactive shifts in neuronal activity (isoflurane anesthesia and CaMKIIa Gi DREADD activation) also increased microglial process calcium signaling. Under hypoactive neuronal conditions, microglia also exhibited process extension and outgrowth with greater calcium signaling. Our work reveals that microglia have highly distinct microdomain signaling, and that processes specifically respond to bi-directional shifts in neuronal activity through increased calcium signaling.
Collapse
Affiliation(s)
| | | | - Yanlu Ying
- Department of Neurology, Mayo Clinic, Rochester, United States
| | - Yong U Liu
- Department of Neurology, Mayo Clinic, Rochester, United States
| | - Gregory Worrell
- Department of Neurology, Mayo Clinic, Rochester, United States
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, United States.,Department of Neuroscience, Mayo Clinic, Jacksonville, United States.,Department of Immunology, Mayo Clinic, Rochester, United States
| |
Collapse
|
42
|
Pagani F, Testi C, Grimaldi A, Corsi G, Cortese B, Basilico B, Baiocco P, De Panfilis S, Ragozzino D, Di Angelantonio S. Dimethyl Fumarate Reduces Microglia Functional Response to Tissue Damage and Favors Brain Iron Homeostasis. Neuroscience 2020; 439:241-254. [DOI: 10.1016/j.neuroscience.2019.10.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 10/08/2019] [Accepted: 10/23/2019] [Indexed: 01/20/2023]
|
43
|
In vivo characterization of functional states of cortical microglia during peripheral inflammation. Brain Behav Immun 2020; 87:243-255. [PMID: 31837418 DOI: 10.1016/j.bbi.2019.12.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022] Open
Abstract
Peripheral inflammation is known to trigger a mirror inflammatory response in the brain, involving brain's innate immune cells - microglia. However, the functional phenotypes, which these cells adopt in the course of peripheral inflammation, remain obscure. In vivo two-photon imaging of microglial Ca2+ signaling as well as process motility reveals two distinct functional states of cortical microglia during a lipopolysaccharide-induced peripheral inflammation: an early "sensor state" characterized by dramatically increased intracellular Ca2+ signaling but ramified morphology and a later "effector state" characterized by slow normalization of intracellular Ca2+ signaling but hypertrophic morphology, substantial IL-1β production in a subset of cells as well as increased velocity of directed process extension and loss of coordination between individual processes. Thus, lipopolysaccharide-induced microglial Ca2+ signaling might represent the central element connecting receptive and executive functions of microglia.
Collapse
|
44
|
Olmedillas Del Moral M, Fröhlich N, Figarella K, Mojtahedi N, Garaschuk O. Effect of Caloric Restriction on the in vivo Functional Properties of Aging Microglia. Front Immunol 2020; 11:750. [PMID: 32411143 PMCID: PMC7198715 DOI: 10.3389/fimmu.2020.00750] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/02/2020] [Indexed: 12/29/2022] Open
Abstract
Throughout the lifespan, microglia, the primary innate immune cells of the brain, fulfill a plethora of homeostatic as well as active immune defense functions, and their aging-induced dysfunctionality is now considered as a key trigger of aging-related brain disorders. Recent evidence suggests that both organism’s sex and age critically impact the functional state of microglia but in vivo determinants of such state(s) remain unclear. Therefore, we analyzed in vivo the sex-specific functional states of microglia in young adult, middle aged and old wild type mice by means of multicolor two-photon imaging, using the microglial Ca2 + signaling and directed process motility as main readouts. Our data revealed the sex-specific differences in microglial Ca2 + signaling at all ages tested, beginning with young adults. Furthermore, for both sexes it showed that during the lifespan the functional state of microglia changes at least twice. Already at middle age the cells are found in the reactive or immune alerted state, characterized by heightened Ca2 + signaling but normal process motility whereas old mice harbor senescent microglia with decreased Ca2 + signaling, and faster but disorganized directed movement of microglial processes. The 6–12 months long caloric restriction (70% of ad libitum food intake) counteracted these aging-induced changes shifting many but not all functional properties of microglia toward a younger phenotype. The improvement of Ca2 + signaling was more pronounced in males. Importantly, even short-term (6-week-long) caloric restriction beginning at old age strongly improved microglial process motility and induced a significant albeit weaker improvement of microglial Ca2 + signaling. Together, these data provide first sex-specific in vivo characterization of functional properties of microglia along the lifespan and identify caloric restriction as a potent, cost-effective, and clinically relevant tool for rejuvenation of microglia.
Collapse
Affiliation(s)
- Maria Olmedillas Del Moral
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nicole Fröhlich
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Katherine Figarella
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Nima Mojtahedi
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
45
|
Figarella K, Wolburg H, Garaschuk O, Duszenko M. Microglia in neuropathology caused by protozoan parasites. Biol Rev Camb Philos Soc 2019; 95:333-349. [PMID: 31682077 DOI: 10.1111/brv.12566] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/31/2022]
Abstract
Involvement of the central nervous system (CNS) is the most severe consequence of some parasitic infections. Protozoal infections comprise a group of diseases that together affect billions of people worldwide and, according to the World Health Organization, are responsible for more than 500000 deaths annually. They include African and American trypanosomiasis, leishmaniasis, malaria, toxoplasmosis, and amoebiasis. Mechanisms underlying invasion of the brain parenchyma by protozoa are not well understood and may depend on parasite nature: a vascular invasion route is most common. Immunosuppression favors parasite invasion into the CNS and therefore the host immune response plays a pivotal role in the development of a neuropathology in these infectious diseases. In the brain, microglia are the resident immune cells active in defense against pathogens that target the CNS. Beside their direct role in innate immunity, they also play a principal role in coordinating the trafficking and recruitment of other immune cells from the periphery to the CNS. Despite their evident involvement in the neuropathology of protozoan infections, little attention has given to microglia-parasite interactions. This review describes the most prominent features of microglial cells and protozoan parasites and summarizes the most recent information regarding the reaction of microglial cells to parasitic infections. We highlight the involvement of the periphery-brain axis and emphasize possible scenarios for microglia-parasite interactions.
Collapse
Affiliation(s)
- Katherine Figarella
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Hartwig Wolburg
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Michael Duszenko
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
46
|
Healthy Brain Aging Modifies Microglial Calcium Signaling In Vivo. Int J Mol Sci 2019; 20:ijms20030589. [PMID: 30704036 PMCID: PMC6386999 DOI: 10.3390/ijms20030589] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/11/2019] [Accepted: 01/24/2019] [Indexed: 12/22/2022] Open
Abstract
Brain aging is characterized by a chronic, low-grade inflammatory state, promoting deficits in cognition and the development of age-related neurodegenerative diseases. Malfunction of microglia, the brain-resident immune cells, was suggested to play a critical role in neuroinflammation, but the mechanisms underlying this malfunctional phenotype remain unclear. Specifically, the age-related changes in microglial Ca2+ signaling, known to be linked to its executive functions, are not well understood. Here, using in vivo two-photon imaging, we characterize intracellular Ca2+ signaling and process extension of cortical microglia in young adult (2–4-month-old), middle-aged (9–11-month-old), and old (18–21-month-old) mice. Our data revealed a complex and nonlinear dependency of the properties of intracellular Ca2+ signals on an animal’s age. While the fraction of cells displaying spontaneous Ca2+ transients progressively increased with age, the frequencies and durations of the spontaneous Ca2+ transients followed a bell-shaped relationship, with the most frequent and largest Ca2+ transients seen in middle-aged mice. Moreover, in old mice microglial processes extending toward an ATP source moved faster but in a more disorganized manner, compared to young adult mice. Altogether, these findings identify two distinct phenotypes of aging microglia: a reactive phenotype, abundantly present in middle-aged animals, and a dysfunctional/senescent phenotype ubiquitous in old mice.
Collapse
|
47
|
Izquierdo P, Attwell D, Madry C. Ion Channels and Receptors as Determinants of Microglial Function. Trends Neurosci 2019; 42:278-292. [PMID: 30678990 DOI: 10.1016/j.tins.2018.12.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022]
Abstract
Microglia provide immune surveillance of the CNS. They display diverse behaviors, including nondirectional and directed motility of their processes, phagocytosis of targets such as dying neurons or superfluous synapses, and generation of reactive oxygen species (ROS) and cytokines. Many of these functions are mediated by ion channels and cell surface receptors, the expression of which varies with the many morphological and functional states that microglial cells can adopt. Recent progress in understanding microglial function has been facilitated by applying classical cell physiological techniques in situ, such as patch-clamping and live imaging, and cell-specific transcriptomic analyses. Here, we review the contribution of microglial ion channels and receptors to microglial and brain function.
Collapse
Affiliation(s)
- Pablo Izquierdo
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower St, London, WC1E 6BT, UK
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower St, London, WC1E 6BT, UK.
| | - Christian Madry
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117 Berlin, Germany.
| |
Collapse
|
48
|
Brawek B, Garaschuk O. Single-Cell Electroporation for Measuring In Vivo Calcium Dynamics in Microglia. Methods Mol Biol 2019; 2034:231-241. [PMID: 31392689 DOI: 10.1007/978-1-4939-9658-2_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Similar to many nonexcitable cells, microglia utilize intracellular Ca2+ signaling for the communication with each other as well as neurons and astrocytes and for triggering a magnitude of their executive functions. However, in vivo measurements of the intracellular Ca2+ dynamics in microglia have been challenging due to technical reasons. Here, we describe an approach utilizing a single-cell electroporation technique to facilitate the study of microglial Ca2+ signaling in the living brain.
Collapse
Affiliation(s)
- Bianca Brawek
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| |
Collapse
|
49
|
Abstract
Microglial cells derive from fetal macrophages which immigrate into and disseminate throughout the central nervous system (CNS) in early embryogenesis. After settling in the nerve tissue, microglial progenitors acquire an idiosyncratic morphological phenotype with small cell body and moving thin and highly ramified processes currently defined as "resting or surveillant microglia". Physiology of microglia is manifested by second messenger-mediated cellular excitability, low resting membrane conductance, and expression of receptors to pathogen- or damage-associated molecular patterns (PAMPs and DAMPs), as well as receptors to classical neurotransmitters and neurohormones. This specific physiological profile reflects adaptive changes of myeloid cells to the CNS environment.
Collapse
Affiliation(s)
- Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
50
|
Tvrdik P, Kearns KN, Sharifi KA, Sluzewski MF, Acton ST, Kalani MYS. Calcium Imaging of Microglial Network Activity in Stroke. Methods Mol Biol 2019; 2034:267-279. [PMID: 31392691 DOI: 10.1007/978-1-4939-9658-2_19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Calcium signaling plays a significant role in microglial activation. Genetically encoded calcium indicators (GECI) have been widely used for calcium imaging studies in many brain cell types, including neurons, astrocytes, and oligodendrocytes. However, microglial calcium imaging approaches have been hampered by idiosyncrasies of their gene expression and malleable cell properties. The generation of PC::G5-tdT, a Polr2a locus-based conditional mouse reporter of calcium, facilitated the deployment of GECI in microglia. When crossed with the Iba1(Aif1)-IRES-Cre line, all brain microglia of the progeny are labeled with the calcium indicator variant GCaMP5G and the red fluorescent protein tdTomato. This reporter system has enabled in vivo studies of intracellular calcium in large microglial cell populations in cerebral pathologies such as ischemic stroke. In this chapter, we outline specific guidelines for genetic, surgical, imaging, and data analysis aspects of microglial calcium monitoring of the ischemic cortex following middle cerebral artery occlusion.
Collapse
Affiliation(s)
- Petr Tvrdik
- Department of Neurosurgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Kathryn N Kearns
- Department of Neurosurgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Khadijeh A Sharifi
- Department of Neurosurgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - M Filip Sluzewski
- Department of Electrical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Scott T Acton
- Department of Electrical Engineering, University of Virginia, Charlottesville, VA, USA
| | - M Yashar S Kalani
- Department of Neurosurgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|