1
|
Wang H, Yang J, Sun Z, Nie Y, He Y. Neoprzewaquinone A alters the migration, phagocytosis and energy metabolism of IL-15-induced HMC3 cells. Mol Immunol 2024; 174:11-17. [PMID: 39128414 DOI: 10.1016/j.molimm.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024]
Abstract
Microglia play a major role in the immune defense system of the central nervous system and are activated in many neurological diseases. The immunomodulatory cytokine interleukin (IL)-15 is known to be involved in microglia response and inflammatory factors release. Neoprzewaquinone A (NEO) is an active compound isolated from Salvia miltiorrhiza Bunge. Our previous study has shown that NEO significantly inhibit the proliferation of IL-15-treated Mo7e cells. However, the role of NEO in the structure and function of IL-15-treated human microglial cells (HMC3) remains unclear. Thus, our study aimed to quantitatively analyze the beneficial effects of NEO on HMC3 cells following IL-15 treatment. The cell viability, phagocytosis, migration and energy metabolism were evaluated by Cell Counting Kit-8 (CCK8), scratch assay, pHrodo™ Red Zymosan BioParticles™ Conjugate, and Agilent Seahorse XF Cell Mito Test. Cephalothin (CEP) was selected as a positive drug because it has obvious inhibitory effect on IL-15 and IL-15Rɑ. Our results showed that IL-15 stimulated the proliferation, migration and phagocytosis of HMC3 cells in a time-dependent manner. Interestingly, NEO exhibited significant suppressive effects on these IL-15-induced changes, which were even superior to those observed with the CEP. Moreover, IL-15 treatment did not significantly alter energy metabolism, including glycolysis and mitochondrial respiration. NEO and CEP alone effectively reduced glycolysis, non-mitochondrial respiration, basal respiration, ATP turnover, respiration capacity, and H+ leak in HMC3 cells. Furthermore, NEO displayed a partial regulatory effect on mitochondrial function in IL-15-treated HMC3 cells. Our study confirms the effectively inhibition of NEO on IL-15-induced microglial activation and provides valuable insights into the therapeutic prospects of NEO in neuropsychiatric disorders associated with IL-15 and microglia.
Collapse
Affiliation(s)
- Haixia Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Jian Yang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Yadan Nie
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Yi He
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
2
|
Berki P, Cserép C, Környei Z, Pósfai B, Szabadits E, Domonkos A, Kellermayer A, Nyerges M, Wei X, Mody I, Kunihiko A, Beck H, Kaikai H, Ya W, Lénárt N, Wu Z, Jing M, Li Y, Gulyás AI, Dénes Á. Microglia contribute to neuronal synchrony despite endogenous ATP-related phenotypic transformation in acute mouse brain slices. Nat Commun 2024; 15:5402. [PMID: 38926390 PMCID: PMC11208608 DOI: 10.1038/s41467-024-49773-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Acute brain slices represent a workhorse model for studying the central nervous system (CNS) from nanoscale events to complex circuits. While slice preparation inherently involves tissue damage, it is unclear how microglia, the main immune cells and damage sensors of the CNS react to this injury and shape neuronal activity ex vivo. To this end, we investigated microglial phenotypes and contribution to network organization and functioning in acute brain slices. We reveal time-dependent microglial phenotype changes influenced by complex extracellular ATP dynamics through P2Y12R and CX3CR1 signalling, which is sustained for hours in ex vivo mouse brain slices. Downregulation of P2Y12R and changes of microglia-neuron interactions occur in line with alterations in the number of excitatory and inhibitory synapses over time. Importantly, functional microglia modulate synapse sprouting, while microglial dysfunction results in markedly impaired ripple activity both ex vivo and in vivo. Collectively, our data suggest that microglia are modulators of complex neuronal networks with important roles to maintain neuronal network integrity and activity. We suggest that slice preparation can be used to model time-dependent changes of microglia-neuron interactions to reveal how microglia shape neuronal circuits in physiological and pathological conditions.
Collapse
Affiliation(s)
- Péter Berki
- János Szentágothai Doctoral School of Neuroscience, Semmelweis University, Budapest, H-1083, Hungary
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Neuronal Network and Behaviour, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zsuzsanna Környei
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Eszter Szabadits
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Andor Domonkos
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Thalamus Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Anna Kellermayer
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Miklós Nyerges
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Xiaofei Wei
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Istvan Mody
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Araki Kunihiko
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - Heinz Beck
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - He Kaikai
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Wang Ya
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Miao Jing
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Attila I Gulyás
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary.
| |
Collapse
|
3
|
Qiao C, Liu Z, Qie S. The Implications of Microglial Regulation in Neuroplasticity-Dependent Stroke Recovery. Biomolecules 2023; 13:biom13030571. [PMID: 36979506 PMCID: PMC10046452 DOI: 10.3390/biom13030571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Stroke causes varying degrees of neurological deficits, leading to corresponding dysfunctions. There are different therapeutic principles for each stage of pathological development. Neuroprotection is the main treatment in the acute phase, and functional recovery becomes primary in the subacute and chronic phases. Neuroplasticity is considered the basis of functional restoration and neurological rehabilitation after stroke, including the remodeling of dendrites and dendritic spines, axonal sprouting, myelin regeneration, synapse shaping, and neurogenesis. Spatiotemporal development affects the spontaneous rewiring of neural circuits and brain networks. Microglia are resident immune cells in the brain that contribute to homeostasis under physiological conditions. Microglia are activated immediately after stroke, and phenotypic polarization changes and phagocytic function are crucial for regulating focal and global brain inflammation and neurological recovery. We have previously shown that the development of neuroplasticity is spatiotemporally consistent with microglial activation, suggesting that microglia may have a profound impact on neuroplasticity after stroke and may be a key therapeutic target for post-stroke rehabilitation. In this review, we explore the impact of neuroplasticity on post-stroke restoration as well as the functions and mechanisms of microglial activation, polarization, and phagocytosis. This is followed by a summary of microglia-targeted rehabilitative interventions that influence neuroplasticity and promote stroke recovery.
Collapse
Affiliation(s)
- Chenye Qiao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| |
Collapse
|
4
|
Ossola B, Rifat A, Rowland A, Hunter H, Drinkall S, Bender C, Hamlischer M, Teall M, Burley R, Barker DF, Cadwalladr D, Dickson L, Lawrence JMK, Harvey JRM, Lizio M, Xu X, Kavanagh E, Cheung T, Sheardown S, Lawrence CB, Harte M, Brough D, Madry C, Matthews K, Doyle K, Page K, Powell J, Brice NL, Bürli RW, Carlton MB, Dawson LA. Characterisation of C101248: A novel selective THIK-1 channel inhibitor for the modulation of microglial NLRP3-inflammasome. Neuropharmacology 2023; 224:109330. [PMID: 36375694 PMCID: PMC9841576 DOI: 10.1016/j.neuropharm.2022.109330] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Neuroinflammation, specifically the NLRP3 inflammasome cascade, is a common underlying pathological feature of many neurodegenerative diseases. Evidence suggests that NLRP3 activation involves changes in intracellular K+. Nuclear Enriched Transcript Sort Sequencing (NETSseq), which allows for deep sequencing of purified cell types from human post-mortem brain tissue, demonstrated a highly specific expression of the tandem pore domain halothane-inhibited K+ channel 1 (THIK-1) in microglia compared to other glial and neuronal cell types in the human brain. NETSseq also showed a significant increase of THIK-1 in microglia isolated from cortical regions of brains with Alzheimer's disease (AD) relative to control donors. Herein, we report the discovery and pharmacological characterisation of C101248, the first selective small-molecule inhibitor of THIK-1. C101248 showed a concentration-dependent inhibition of both mouse and human THIK-1 (IC50: ∼50 nM) and was inactive against K2P family members TREK-1 and TWIK-2, and Kv2.1. Whole-cell patch-clamp recordings of microglia from mouse hippocampal slices showed that C101248 potently blocked both tonic and ATP-evoked THIK-1 K+ currents. Notably, C101248 had no effect on other constitutively active resting conductance in slices from THIK-1-depleted mice. In isolated microglia, C101248 prevented NLRP3-dependent release of IL-1β, an effect not seen in THIK-1-depleted microglia. In conclusion, we demonstrated that inhibiting THIK-1 (a microglia specific gene that is upregulated in brains from donors with AD) using a novel selective modulator attenuates the NLRP3-dependent release of IL-1β from microglia, which suggests that this channel may be a potential therapeutic target for the modulation of neuroinflammation in AD.
Collapse
Affiliation(s)
- Bernardino Ossola
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK.
| | - Ali Rifat
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Anna Rowland
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Helen Hunter
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Samuel Drinkall
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Clare Bender
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Mayida Hamlischer
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Martin Teall
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Russell Burley
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Daneil F Barker
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - David Cadwalladr
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Louise Dickson
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Jason M K Lawrence
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Jenna R M Harvey
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Marina Lizio
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Xiao Xu
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Edel Kavanagh
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Toni Cheung
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Steve Sheardown
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Catherine B Lawrence
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Michael Harte
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - David Brough
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Christian Madry
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Kim Matthews
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Kevin Doyle
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Keith Page
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Justin Powell
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Nicola L Brice
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Roland W Bürli
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Mark B Carlton
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Lee A Dawson
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| |
Collapse
|
5
|
Gonçalves de Andrade E, Šimončičová E, Carrier M, Vecchiarelli HA, Robert MÈ, Tremblay MÈ. Microglia Fighting for Neurological and Mental Health: On the Central Nervous System Frontline of COVID-19 Pandemic. Front Cell Neurosci 2021; 15:647378. [PMID: 33737867 PMCID: PMC7961561 DOI: 10.3389/fncel.2021.647378] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is marked by cardio-respiratory alterations, with increasing reports also indicating neurological and psychiatric symptoms in infected individuals. During COVID-19 pathology, the central nervous system (CNS) is possibly affected by direct severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) invasion, exaggerated systemic inflammatory responses, or hypoxia. Psychosocial stress imposed by the pandemic further affects the CNS of COVID-19 patients, but also the non-infected population, potentially contributing to the emergence or exacerbation of various neurological or mental health disorders. Microglia are central players of the CNS homeostasis maintenance and inflammatory response that exert their crucial functions in coordination with other CNS cells. During homeostatic challenges to the brain parenchyma, microglia modify their density, morphology, and molecular signature, resulting in the adjustment of their functions. In this review, we discuss how microglia may be involved in the neuroprotective and neurotoxic responses against CNS insults deriving from COVID-19. We examine how these responses may explain, at least partially, the neurological and psychiatric manifestations reported in COVID-19 patients and the general population. Furthermore, we consider how microglia might contribute to increased CNS vulnerability in certain groups, such as aged individuals and people with pre-existing conditions.
Collapse
Affiliation(s)
| | - Eva Šimončičová
- Division of Medical Science, University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Division of Medical Science, University of Victoria, Victoria, BC, Canada.,Axe Neurosciences, Centre de Recherche du CHU de Québec, Université de Laval, Québec City, QC, Canada
| | | | - Marie-Ève Robert
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université de Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Science, University of Victoria, Victoria, BC, Canada.,Axe Neurosciences, Centre de Recherche du CHU de Québec, Université de Laval, Québec City, QC, Canada.,Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada.,Department of Molecular Medicine, Université de Laval, Québec City, QC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Dissection of P2X4 and P2X7 Receptor Current Components in BV-2 Microglia. Int J Mol Sci 2020; 21:ijms21228489. [PMID: 33187309 PMCID: PMC7696836 DOI: 10.3390/ijms21228489] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
Microglia cells represent the immune system of the central nervous system. They become activated by ATP released from damaged and inflamed tissue via purinergic receptors. Ionotropic purinergic P2X4 and P2X7 receptors have been shown to be involved in neurological inflammation and pain sensation. Whether the two receptors assemble exclusively as homotrimers or also as heterotrimers is still a matter of debate. We investigated the expression of P2X receptors in BV-2 microglia cells applying the whole-cell voltage-clamp technique. We dissected P2X4 and P2X7 receptor-mediated current components by using specific P2X4 and P2X7 receptor blockers and by their characteristic current kinetics. We found that P2X4 and P2X7 receptors are activated independently from each other, indicating that P2X4/P2X7 heteromers are not of functional significance in these cells. The pro-inflammatory mediators lipopolysaccharide and interferon γ, if applied in combination, upregulated P2X4, but not P2X7 receptor-dependent current components also arguing against phenotypically relevant heteromerization of P2X4 and P2X7 receptor subunits.
Collapse
|
7
|
Onodera J, Nagata H, Nakashima A, Ikegaya Y, Koyama R. Neuronal brain-derived neurotrophic factor manipulates microglial dynamics. Glia 2020; 69:890-904. [PMID: 33119934 DOI: 10.1002/glia.23934] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 10/22/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
Abstract
Brain-derived neurotrophic factor (BDNF), a main member of the neurotrophin family that is active in the brain, supports neuronal survival and growth. Microglial BDNF affects both the structural and functional properties of neurons. In contrast, whether and how neuronal BDNF affects microglial dynamics remain largely undetermined. Here, we examined the effects of BDNF on the properties of microglia in the CA3 region of the hippocampus. We chose this site because the axonal boutons of hippocampal mossy fibers, which are mostly formed in the CA3 region, contain the highest levels of BDNF in the rodent brain. We transfected mouse dentate granule cells with an adeno-associated virus that encodes both a BDNF short hairpin RNA (shRNA) and red fluorescent protein to examine the effects of mossy fiber-derived BDNF on microglia. Based on immunohistochemistry, BDNF knockdown with an shRNA resulted in an increase in microglial density in the mossy fiber pathway and increased engulfment of mossy fiber axons by microglia. In addition, we performed time-lapse imaging of microglial processes in hippocampal slice cultures to examine the effects of BDNF on microglial motility. Time-lapse imaging revealed increases in the motility of microglial processes and the engulfment of mossy fiber synapses by microglia when BDNF signaling was pharmacologically blocked. Thus, neuronal BDNF prevents microglia from engulfing mossy fiber synapses in the hippocampus.
Collapse
Affiliation(s)
- Junya Onodera
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidetaka Nagata
- Platform Technology Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Ai Nakashima
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuji Ikegaya
- 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
|
8
|
Kluge MG, Abdolhoseini M, Zalewska K, Ong LK, Johnson SJ, Nilsson M, Walker FR. Spatiotemporal analysis of impaired microglia process movement at sites of secondary neurodegeneration post-stroke. J Cereb Blood Flow Metab 2019; 39:2456-2470. [PMID: 30204044 PMCID: PMC6893987 DOI: 10.1177/0271678x18797346] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It has recently been identified that after motor cortex stroke, the ability of microglia processes to respond to local damage cues is lost from the thalamus, a major site of secondary neurodegeneration (SND). In this study, we combine a photothrombotic stroke model in mice, acute slice and fluorescent imaging to analyse the loss of microglia process responsiveness. The peri-infarct territories and thalamic areas of SND were investigated at time-points 3, 7, 14, 28 and 56 days after stroke. We confirmed the highly specific nature of non-responsive microglia processes to sites of SND. Non-responsiveness was at no time observed at the peri-infarct but started in the thalamus seven days post-stroke and persisted for 56 days. Loss of directed process extension is not a reflection of general functional paralysis as phagocytic function continued to increase over time. Additionally, we identified that somal P2Y12 was present on non-responsive microglia in the first two weeks after stroke but not at later time points. Finally, both classical microglia activation and loss of process extension are highly correlated with neuronal damage. Our findings highlight the importance of microglia, specifically microglia dynamic functions, to the progression of SND post-stroke, and their potential relevance as modulators or therapeutic targets during stroke recovery.
Collapse
Affiliation(s)
- Murielle G Kluge
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Mahmoud Abdolhoseini
- School of Electrical Engineering and Computer Science, University of Newcastle, Callaghan, NSW, Australia
| | - Katarzyna Zalewska
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Lin Kooi Ong
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia
| | - Sarah J Johnson
- School of Electrical Engineering and Computer Science, University of Newcastle, Callaghan, NSW, Australia
| | - Michael Nilsson
- Hunter Medical Research Institute, Newcastle, NSW, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia
| |
Collapse
|
9
|
Eyo UB, Wu LJ. Microglia: Lifelong patrolling immune cells of the brain. Prog Neurobiol 2019; 179:101614. [PMID: 31075285 PMCID: PMC6599472 DOI: 10.1016/j.pneurobio.2019.04.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/11/2019] [Accepted: 04/19/2019] [Indexed: 02/02/2023]
Abstract
Microglial cells are the predominant parenchymal immune cell of the brain. Recent evidence suggests that like peripheral immune cells, microglia patrol the brain in health and disease. Reviewing these data, we first examine the evidence that microglia invade the brain mesenchyme early in embryonic development, establish residence therein, proliferate and subsequently maintain their numbers throughout life. We, then, summarize established and novel evidence for microglial process surveillance in the healthy and injured brain. Finally, we discuss emerging evidence for microglial cell body dynamics that challenge existing assumptions of their sessile nature. We conclude that microglia are long-lived immune cells that patrol the brain through both cell body and process movements. This recognition has significant implications for neuroimmune interactions throughout the animal lifespan.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
10
|
The Evolving Dialogue of Microglia and Neurons in Alzheimer’s Disease: Microglia as Necessary Transducers of Pathology. Neuroscience 2019; 405:24-34. [DOI: 10.1016/j.neuroscience.2018.01.059] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 01/21/2023]
|
11
|
Smolders SMT, Kessels S, Vangansewinkel T, Rigo JM, Legendre P, Brône B. Microglia: Brain cells on the move. Prog Neurobiol 2019; 178:101612. [PMID: 30954517 DOI: 10.1016/j.pneurobio.2019.04.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/13/2019] [Accepted: 04/01/2019] [Indexed: 02/08/2023]
Abstract
In the last decade, tremendous progress has been made in understanding the biology of microglia - i.e. the fascinating immigrated resident immune cell population of the central nervous system (CNS). Recent literature reviews have largely dealt with the plentiful functions of microglia in CNS homeostasis, development and pathology, and the influences of sex and the microbiome. In this review, the intriguing aspect of their physical plasticity during CNS development will get specific attention. Microglia move around (mobility) and reshape their processes (motility). Microglial migration into and inside the CNS is most prominent throughout development and consequently most of the data described in this review concern mobility and motility in the changing environment of the developing brain. Here, we first define microglia based on their highly specialized age- and region-dependent gene expression signature and associated functional heterogeneity. Next, we describe their origin, the migration route of immature microglial cells towards the CNS, the mechanisms underlying their invasion of the CNS, and their spatiotemporal localization and surveying behaviour inside the developing CNS. These processes are dependent on microglial mobility and motility which are determined by the microenvironment of the CNS. Therefore, we further zoom in on the changing environment during CNS development. We elaborate on the extracellular matrix and the respective integrin receptors on microglia and we discuss the purinergic and molecular signalling in microglial mobility. In the last section, we discuss the physiological and pathological functions of microglia in which mobility and motility are involved to stress the importance of microglial 'movement'.
Collapse
Affiliation(s)
- Sophie Marie-Thérèse Smolders
- UHasselt, BIOMED, Diepenbeek, Belgium; INSERM, UMR-S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France; Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | | | | | | - Pascal Legendre
- INSERM, UMR-S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France; Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | |
Collapse
|
12
|
Kyrargyri V, Attwell D, Jolivet RB, Madry C. Analysis of Signaling Mechanisms Regulating Microglial Process Movement. Methods Mol Biol 2019; 2034:191-205. [PMID: 31392686 DOI: 10.1007/978-1-4939-9658-2_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Microglia, the brain's innate immune cells, are extremely motile cells, continuously surveying the central nervous system (CNS) to serve homeostatic functions and to respond to pathological events. In the healthy brain, microglia exhibit a small cell body with long, branched, and highly motile processes, which constantly extend and retract, effectively "patrolling" the brain parenchyma. Over the last decade, methodological advances in microscopy and the availability of genetically encoded reporter mice have allowed us to probe microglial physiology in situ. Beyond their classical immunological roles, unexpected functions of microglia have been revealed, both in the developing and the adult brain: microglia regulate the generation of newborn neurons, control the formation and elimination of synapses, and modulate neuronal activity. Many of these newly ascribed functions depend directly on microglial process movement. Thus, elucidating the mechanisms underlying microglial motility is of great importance to understand their role in brain physiology and pathophysiology. Two-photon imaging of fluorescently labeled microglia, either in vivo or ex vivo in acute brain slices, has emerged as an indispensable tool for investigating microglial movements and their functional consequences. This chapter aims to provide a detailed description of the experimental data acquisition and analysis needed to address these questions, with a special focus on key dynamic and morphological metrics such as surveillance, directed motility, and ramification.
Collapse
Affiliation(s)
- Vasiliki Kyrargyri
- Department of Immunology, Laboratory of Molecular Genetics, Hellenic Pasteur Institute, Athens, Greece
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Renaud Blaise Jolivet
- Département de Physique Nucléaire et Corpusculaire, University of Geneva, Geneva, Switzerland
- CERN, Geneva, Switzerland
| | - Christian Madry
- Institute of Neurophysiology, Charité-Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
13
|
Madry C, Arancibia-Cárcamo IL, Kyrargyri V, Chan VTT, Hamilton NB, Attwell D. Effects of the ecto-ATPase apyrase on microglial ramification and surveillance reflect cell depolarization, not ATP depletion. Proc Natl Acad Sci U S A 2018; 115:E1608-E1617. [PMID: 29382767 PMCID: PMC5816168 DOI: 10.1073/pnas.1715354115] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Microglia, the brain's innate immune cells, have highly motile processes which constantly survey the brain to detect infection, remove dying cells, and prune synapses during brain development. ATP released by tissue damage is known to attract microglial processes, but it is controversial whether an ambient level of ATP is needed to promote constant microglial surveillance in the normal brain. Applying the ATPase apyrase, an enzyme which hydrolyzes ATP and ADP, reduces microglial process ramification and surveillance, suggesting that ambient ATP/ADP maintains microglial surveillance. However, attempting to raise the level of ATP/ADP by blocking the endogenous ecto-ATPase (termed NTPDase1/CD39), which also hydrolyzes ATP/ADP, does not affect the cells' ramification or surveillance, nor their membrane currents, which respond to even small rises of extracellular [ATP] or [ADP] with the activation of K+ channels. This indicates a lack of detectable ambient ATP/ADP and ecto-ATPase activity, contradicting the results with apyrase. We resolve this contradiction by demonstrating that contamination of commercially available apyrase by a high K+ concentration reduces ramification and surveillance by depolarizing microglia. Exposure to the same K+ concentration (without apyrase added) reduced ramification and surveillance as with apyrase. Dialysis of apyrase to remove K+ retained its ATP-hydrolyzing activity but abolished the microglial depolarization and decrease of ramification produced by the undialyzed enzyme. Thus, applying apyrase affects microglia by an action independent of ATP, and no ambient purinergic signaling is required to maintain microglial ramification and surveillance. These results also have implications for hundreds of prior studies that employed apyrase to hydrolyze ATP/ADP.
Collapse
Affiliation(s)
- Christian Madry
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom;
- Institute of Neurophysiology, Charité Universitätsmedizin, 10117 Berlin, Germany
| | - I Lorena Arancibia-Cárcamo
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Vasiliki Kyrargyri
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Victor T T Chan
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Nicola B Hamilton
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom;
- Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom;
| |
Collapse
|
14
|
Roesch S, Rapp C, Dettling S, Herold-Mende C. When Immune Cells Turn Bad-Tumor-Associated Microglia/Macrophages in Glioma. Int J Mol Sci 2018; 19:ijms19020436. [PMID: 29389898 PMCID: PMC5855658 DOI: 10.3390/ijms19020436] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 12/29/2017] [Accepted: 01/29/2018] [Indexed: 12/31/2022] Open
Abstract
As a substantial part of the brain tumor microenvironment (TME), glioma-associated microglia/macrophages (GAMs) have an emerging role in tumor progression and in controlling anti-tumor immune responses. We review challenges and improvements of cell models and highlight the contribution of this highly plastic cell population to an immunosuppressive TME, besides their well-known functional role regarding glioma cell invasion and angiogenesis. Finally, we summarize first therapeutic interventions to target GAMs and their effect on the immunobiology of gliomas, focusing on their interaction with T cells.
Collapse
Affiliation(s)
- Saskia Roesch
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany.
| | - Carmen Rapp
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany.
| | - Steffen Dettling
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany.
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany.
| |
Collapse
|
15
|
The GluN2A Subunit Regulates Neuronal NMDA receptor-Induced Microglia-Neuron Physical Interactions. Sci Rep 2018; 8:828. [PMID: 29339791 PMCID: PMC5770428 DOI: 10.1038/s41598-018-19205-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 12/19/2017] [Indexed: 11/11/2022] Open
Abstract
Microglia are known to engage in physical interactions with neurons. However, our understanding of the detailed mechanistic regulation of microglia-neuron interactions is incomplete. Here, using high resolution two photon imaging, we investigated the regulation of NMDA receptor-induced microglia-neuron physical interactions. We found that the GluN2A inhibitor NVPAAM007, but not the GluN2B inhibitor ifenprodil, blocked the occurrence of these interactions. Consistent with the well-known developmental regulation of the GluN2A subunit, these interactions are absent in neonatal tissues. Furthermore, consistent with a preferential synaptic localization of GluN2A subunits, there is a differential sensitivity of their occurrence between denser (stratum radiatum) and less dense (stratum pyramidale) synaptic sub-regions of the CA1. Finally, consistent with differentially expressed GluN2A subunits in the CA1 and DG areas of the hippocampus, these interactions could not be elicited in the DG despite robust microglial chemotactic capabilities. Together, these results enhance our understanding of the mechanistic regulation of NMDA receptor-dependent microglia-neuronal physical interactions phenomena by the GluN2A subunit that may be relevant in the mammalian brain during heightened glutamatergic neurotransmission such as epilepsy and ischemic stroke.
Collapse
|
16
|
Tvrdik P, Kalani MYS. In Vivo Imaging of Microglial Calcium Signaling in Brain Inflammation and Injury. Int J Mol Sci 2017; 18:ijms18112366. [PMID: 29117112 PMCID: PMC5713335 DOI: 10.3390/ijms18112366] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/01/2017] [Accepted: 11/04/2017] [Indexed: 12/20/2022] Open
Abstract
Microglia, the innate immune sentinels of the central nervous system, are the most dynamic cells in the brain parenchyma. They are the first responders to insult and mediate neuroinflammation. Following cellular damage, microglia extend their processes towards the lesion, modify their morphology, release cytokines and other mediators, and eventually migrate towards the damaged area and remove cellular debris by phagocytosis. Intracellular Ca2+ signaling plays important roles in many of these functions. However, Ca2+ in microglia has not been systematically studied in vivo. Here we review recent findings using genetically encoded Ca2+ indicators and two-photon imaging, which have enabled new insights into Ca2+ dynamics and signaling pathways in large populations of microglia in vivo. These new approaches will help to evaluate pre-clinical interventions and immunomodulation for pathological brain conditions such as stroke and neurodegenerative diseases.
Collapse
Affiliation(s)
- Petr Tvrdik
- Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - M Yashar S Kalani
- Department of Neurosurgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
17
|
Kaur C, Rathnasamy G, Ling EA. Biology of Microglia in the Developing Brain. J Neuropathol Exp Neurol 2017; 76:736-753. [PMID: 28859332 DOI: 10.1093/jnen/nlx056] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Microglia exist in different morphological forms in the developing brain. They show a small cell body with scanty cytoplasm with many branching processes in the grey matter of the developing brain. However, in the white matter such as the corpus callosum where the unmyelinated axons are loosely organized, they appear in an amoeboid form having a round cell body endowed with copious cytoplasm rich in organelles. The amoeboid cells eventually transform into ramified microglia in the second postnatal week when the tissue becomes more compact with the onset of myelination. Microglia serve as immunocompetent macrophages that act as neuropathology sensors to detect and respond swiftly to subtle changes in the brain tissues in pathological conditions. Microglial functions are broadly considered as protective in the normal brain development as they phagocytose dead cells and sculpt neuronal connections by pruning excess axons and synapses. They also secrete a number of trophic factors such as insulin-like growth factor-1 and transforming growth factor-β among many others that are involved in neuronal and oligodendrocyte survival. On the other hand, microglial cells when activated produce a plethora of molecules such as proinflammatory cytokines, chemokines, reactive oxygen species, and nitric oxide that are implicated in the pathogenesis of many pathological conditions such as epilepsy, cerebral palsy, autism, and perinatal hypoxic-ischemic brain injury. Although many studies have investigated the origin and functions of the microglia in the developing brain, in-depth in vivo studies along with analysis of their transcriptome and epigenetic changes need to be undertaken to elucidate their full potential be it protective or neurotoxic. This would lead to a better understanding of their roles in the healthy and diseased developing brain and advancement of therapeutic strategies to target microglia-mediated neurotoxicity.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Gurugirijha Rathnasamy
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
18
|
Purine Signaling and Microglial Wrapping. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 949:147-165. [PMID: 27714688 DOI: 10.1007/978-3-319-40764-7_7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Microglial cells are highly dynamic cells with processes continuously moving to survey the surrounding territory. Microglia possess a broad variety of surface receptors and subtle changes in their microenvironment cause microglial cell processes to extend, retract, and interact with neuronal synaptic contacts. When the nervous system is disturbed, microglia activate, proliferate, and migrate to sites of injury in response to alert signals. Released nucleotides like ATP and UTP are among the wide range of molecules promoting microglial activation and guiding their migration and phagocytic function. The increased concentration of nucleotides in the extracellular space could be involved in the microglial wrapping found around injured neurons in various pathological conditions, especially after peripheral axotomy. Microglial wrappings isolate injured neurons from synaptic inputs and facilitate the molecular dialog between endangered or injured neurons and activated microglia. Astrocytes may also participate in neuronal ensheathment. Degradation of ATP by microglial ecto-nucleotidases and the expression of various purine receptors might be decisive in regulating the function of enwrapping glial cells and in determining the fate of damaged neurons, which may die or may regenerate their axons and survive.
Collapse
|
19
|
Martín-Estebané M, Navascués J, Sierra-Martín A, Martín-Guerrero SM, Cuadros MA, Carrasco MC, Marín-Teva JL. Onset of microglial entry into developing quail retina coincides with increased expression of active caspase-3 and is mediated by extracellular ATP and UDP. PLoS One 2017; 12:e0182450. [PMID: 28763502 PMCID: PMC5538646 DOI: 10.1371/journal.pone.0182450] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 07/18/2017] [Indexed: 12/31/2022] Open
Abstract
Microglial cell precursors located in the area of the base of the pecten and the optic nerve head (BP/ONH) start to enter the retina of quail embryos at the 7th day of incubation (E7), subsequently colonizing the entire retina by central-to-peripheral tangential migration, as previously shown by our group. The present study demonstrates a precise chronological coincidence of the onset of microglial cell entry into the retina with a striking increase in death of retinal cells, as revealed by their active caspase-3 expression and TUNEL staining, in regions dorsal to the BP/ONH area, suggesting that dying retinal cells would contribute to the microglial cell inflow into the retina. However, the molecular mechanisms involved in this inflow are currently unclear. Extracellular nucleotides, such as ATP and UDP, have previously been shown to favor migration of microglia towards brain injuries because they are released by apoptotic cells and stimulate both chemotaxis and chemokinesis in microglial cells via signaling through purinergic receptors. Hence, we tested here the hypothesis that ATP and UDP play a role in the entry and migration of microglial precursors into the developing retina. For this purpose, we used an experimental model system based on organotypic cultures of E6.5 quail embryo retina explants, which mimics the entry and migration of microglial precursors in the in situ developing retina. Inhibition of purinergic signaling by treating retina explants with either apyrase, a nucleotide-hydrolyzing enzyme, or suramin, a broad spectrum antagonist of purinergic receptors, significantly prevents the entry of microglial cells into the retina. In addition, treatment of retina explants with either exogenous ATP or UDP results in significantly increased numbers of microglial cells entering the retina. In light of these findings, we conclude that purinergic signaling by extracellular ATP and UDP is necessary for the entry and migration of microglial cells into the embryonic retina by inducing chemokinesis in these cells.
Collapse
Affiliation(s)
- María Martín-Estebané
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Julio Navascués
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Ana Sierra-Martín
- Departamento de Biología Celular, Fisiología e Inmunología, Facultad de Biociencias, Universidad Autónoma de Barcelona, Bellaterra, Barcelona, Spain
| | | | - Miguel A. Cuadros
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - María-Carmen Carrasco
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - José L. Marín-Teva
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
- * E-mail:
| |
Collapse
|
20
|
Nemeth CL, Drummond GT, Mishra MK, Zhang F, Carr P, Garcia MS, Doman S, Fatemi A, Johnston MV, Kannan RM, Kannan S, Wilson MA. Uptake of dendrimer-drug by different cell types in the hippocampus after hypoxic-ischemic insult in neonatal mice: Effects of injury, microglial activation and hypothermia. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2359-2369. [PMID: 28669854 DOI: 10.1016/j.nano.2017.06.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/15/2017] [Accepted: 06/22/2017] [Indexed: 12/13/2022]
Abstract
Perinatal hypoxic-ischemic encephalopathy (HIE) can result in neurodevelopmental disability, including cerebral palsy. The only treatment, hypothermia, provides incomplete neuroprotection. Hydroxyl polyamidoamine (PAMAM) dendrimers are being explored for targeted delivery of therapy for HIE. Understanding the biodistribution of dendrimer-conjugated drugs into microglia, neurons and astrocytes after brain injury is essential for optimizing drug delivery. We conjugated N-acetyl-L-cysteine to Cy5-labeled PAMAM dendrimer (Cy5-D-NAC) and used a mouse model of perinatal HIE to study effects of timing of administration, hypothermia, brain injury, and microglial activation on uptake. Dendrimer conjugation delivered therapy most effectively to activated microglia but also targeted some astrocytes and injured neurons. Cy5-D-NAC uptake was correlated with brain injury in all cell types and with activated morphology in microglia. Uptake was not inhibited by hypothermia, except in CD68+ microglia. Thus, dendrimer-conjugated drug delivery can target microglia, astrocytes and neurons and can be used in combination with hypothermia for treatment of HIE.
Collapse
Affiliation(s)
- Christina L Nemeth
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA
| | - Gabrielle T Drummond
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA
| | - Manoj K Mishra
- Center for Nanomedicine at the Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA
| | - Fan Zhang
- Center for Nanomedicine at the Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA
| | - Patrice Carr
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA
| | - Maxine S Garcia
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA
| | - Sydney Doman
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA
| | - Ali Fatemi
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA
| | - Michael V Johnston
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA
| | - Rangaramanujam M Kannan
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA; Center for Nanomedicine at the Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 400 N Broadway, Baltimore, MD 21287, USA
| | - Sujatha Kannan
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA; Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, The Charlotte R. Bloomberg Children's Center, 1800 Orleans Street, Suite 6318D, Baltimore, MD 21287, USA.
| | - Mary Ann Wilson
- Hugo W. Moser Research Institute at Kennedy Krieger, 707 N Broadway, Baltimore, MD 21205, USA; Department of Neurology, The Johns Hopkins University School of Medicine, 1800 Orleans St., Baltimore, MD 21287, USA; Department of Neuroscience, The Johns Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205, USA.
| |
Collapse
|
21
|
Cai Q, Li Y, Pei G. Polysaccharides from Ganoderma lucidum attenuate microglia-mediated neuroinflammation and modulate microglial phagocytosis and behavioural response. J Neuroinflammation 2017; 14:63. [PMID: 28340576 PMCID: PMC5364682 DOI: 10.1186/s12974-017-0839-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 03/13/2017] [Indexed: 11/16/2022] Open
Abstract
Background Ganoderma lucidum (GL) has been widely used in Asian countries for hundreds of years to promote health and longevity. The pharmacological functions of which had been classified, including the activation of innate immune responses, suppression of tumour and modulation of cell proliferations. Effective fractions of Ganoderma lucidum polysaccharides (GLP) had already been reported to regulate the immune system. Nevertheless, the role of GLP in the microglia-mediated neuroinflammation has not been sufficiently elucidated. Further, GLP effect on microglial behavioural modulations in correlation with the inflammatory responses remains to be unravelled. The aim of this work was to quantitatively analyse the contributions of GLP on microglia. Methods The BV2 microglia and primary mouse microglia were stimulated by lipopolysaccharides (LPS) and amyloid beta42 (Aβ42) oligomer, respectively. Investigation on the effect of GLP was carried by quantitative determination of the microglial pro- and anti-inflammatory cytokine expressions and behavioural modulations including migration, morphology and phagocytosis. Analysis of microglial morphology and phagocytosis modulations was confirmed in the zebrafish brain. Results Quantitative results revealed that GLP down-regulates LPS- or Aβ-induced pro-inflammatory cytokines and promotes anti-inflammatory cytokine expressions in BV-2 and primary microglia. In addition, GLP attenuates inflammation-related microglial migration, morphological alterations and phagocytosis probabilities. We also showed that modulations of microglial behavioural responses were associated with MCP-1 and C1q expressions. Conclusions Overall, our study provides an insight into the GLP regulation of LPS- and Aβ-induced neuroinflammation and serves an implication that the neuroprotective function of GLP might be achieved through modulation of microglial inflammatory and behavioural responses. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0839-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Cai
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.,Graduate School, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yuanyuan Li
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.,Graduate School, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Gang Pei
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China. .,School of Life Science and Technology, and the Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
22
|
Tozaki-Saitoh H, Miyata H, Yamashita T, Matsushita K, Tsuda M, Inoue K. P2Y12 receptors in primary microglia activate nuclear factor of activated T-cell signaling to induce C-C chemokine 3 expression. J Neurochem 2017; 141:100-110. [PMID: 28144998 DOI: 10.1111/jnc.13968] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 02/03/2023]
Abstract
Microglia are widely accepted as surveillants in the central nervous system that are continually searching the local environment for signs of injury. Following an inflammatory situation, microglia alter their morphology, extend ramified processes, and undergo cell body hypertrophy. Extracellular nucleotides are recognized as a danger signal by microglia. ADP acting on P2Y12 receptors induce process extension of microglia thereby attracting microglia to the site of adenosine tri-phosphate/ADP leaking or release. However, the question whether ADP/P2Y12 receptor signaling directly stimulates the production or release of inducible factors such as cytokines remains unclear. In this study, we found that CC chemokine ligand 3 (CCL3) is induced by ADP-treated primary microglia. Pharmacological characterization using pertussis toxin, a P2Y12 receptor inhibitor, and a calcium chelator revealed that CCL3 induction was caused by P2Y12 receptor-mediated intracellular calcium elevation. Next, nuclear factor of activated T-cell dephosphorylation and nuclear translocalization were observed. Calcineurin, an inhibitor for nuclear factor of activated T cell, suppressed CCL3 induction. These data indicate that microglial P2Y12 receptors are utilized to trigger an acute inflammatory response in microglia via rapid CCL3 induction after ADP stimulation.
Collapse
Affiliation(s)
- Hidetoshi Tozaki-Saitoh
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Miyata
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Yamashita
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuyuki Matsushita
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu Universisty, Fukuoka, Japan
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhide Inoue
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Yang X, Lou Y, Liu G, Wang X, Qian Y, Ding J, Chen S, Xiao Q. Microglia P2Y6 receptor is related to Parkinson's disease through neuroinflammatory process. J Neuroinflammation 2017; 14:38. [PMID: 28219441 PMCID: PMC5319038 DOI: 10.1186/s12974-017-0795-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/11/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Microglia in the central nervous system (CNS) were reported to play crucial role in neurodegeneration. Previous studies showed that P2Y6 receptor (P2Y6R) mainly contributed to microglia activation and phagocytosis in CNS. However, the level of P2Y6R in Parkinson's disease (PD) patients is unclear. Therefore, we measured the level of P2Y6R in PD patients and speculated whether it could be a potential biomarker for PD. Given on the basis that P2Y6R was higher in PD patients, we further explored the mechanisms underlying P2Y6R in the pathogenesis of PD. METHODS We tested the expression level of P2Y6R in the peripheral blood mononuclear cells (PBMCs) among 145 PD patients, 170 healthy controls, and 30 multiple system atrophy (MSA) patients. We also used a lipopolysaccharide (LPS)-stimulated microglial cell culture model to investigate (i) the effects of LPS on P2Y6R expression with western blot and RT-PCR, (ii) the effects of LPS on UDP expression using HPLC, (iii) the effects of UDP/P2Y6R signaling on cytokine expression using western blot, RT-PCR, and ELISA, and (iv) the signaling pathways activated by the P2Y6R involved in the neuroinflammation. RESULTS Expression levels of P2Y6R in PD patients were higher than healthy controls and MSA patients. P2Y6R could be a good biomarker of PD. P2Y6R was also upregulated in LPS-treated BV-2 cells and involved in proinflammatory cytokine release through an autocrine loop based on LPS-triggered UDP secretion and accelerated neuroinflammatory responses through the ERK1/2 pathway. Importantly, blocking UDP/P2Y6R signaling could reverse these pathological processes. CONCLUSIONS P2Y6R may be a potential clinical biomarker of PD. Blocking P2Y6R may be a potential therapeutic approach to the treatment of PD patients through inhibition of microglia-activated neuroinflammation.
Collapse
Affiliation(s)
- Xiaodong Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Yue Lou
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Guidong Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Xueping Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Yiwei Qian
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Jianqing Ding
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| | - Qin Xiao
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
24
|
Liu M, Yao M, Wang H, Xu L, Zheng Y, Huang B, Ni H, Xu S, Zhou X, Lian Q. P2Y 12 receptor-mediated activation of spinal microglia and p38MAPK pathway contribute to cancer-induced bone pain. J Pain Res 2017; 10:417-426. [PMID: 28243146 PMCID: PMC5317303 DOI: 10.2147/jpr.s124326] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Cancer-induced bone pain (CIBP) is one of the most challenging clinical problems due to a lack of understanding the mechanisms. Recent evidence has demonstrated that activation of microglial G-protein-coupled P2Y12 receptor (P2Y12R) and proinflammatory cytokine production play an important role in neuropathic pain generation and maintenance. However, whether P2Y12R is involved in CIBP remains unknown. Methods The purpose of this study was to investigate the role of P2Y12R in CIBP and its molecular mechanisms. Using the bone cancer model inoculated with Walker 256 tumor cells into the left tibia of Sprague Dawley rat, we blocked spinal P2Y12R through intrathecal administration of its selective antagonist MRS2395 (400 pmol/µL, 15 µL). Results We found that not only the ionized calcium-binding adapter molecule 1 (Iba-1)-positive microglia in the ipsilateral spinal cord but also mechanical allodynia was significantly inhibited. Furthermore, it decreased the phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK) and the production of proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-6 (IL-6), whereas it increased tumor necrosis factor-α (TNF-α). Conclusion Taken together, our present results suggest that microglial P2Y12R in the spinal cord may contribute to CIBP by the activation of spinal microglia and p38MAPK pathway, thus identifying a potential therapeutic target for the treatment of CIBP.
Collapse
Affiliation(s)
- Mingjuan Liu
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Ming Yao
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing; Department of Anesthesiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Hanqi Wang
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Longsheng Xu
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Ying Zheng
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Bing Huang
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Huadong Ni
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Shijie Xu
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Xuyan Zhou
- Department of Anesthesiology and Pain Medicine, The First Hospital of Jiaxing, The First Affiliated Hospital of Jiaxing University, Jiaxing
| | - Qingquan Lian
- Department of Anesthesiology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
25
|
Microglial P2Y12 receptors regulate microglial activation and surveillance during neuropathic pain. Brain Behav Immun 2016; 55:82-92. [PMID: 26576724 PMCID: PMC4864135 DOI: 10.1016/j.bbi.2015.11.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/15/2015] [Accepted: 11/09/2015] [Indexed: 11/20/2022] Open
Abstract
Microglial cells are critical in the pathogenesis of neuropathic pain and several microglial receptors have been proposed to mediate this process. Of these receptors, the P2Y12 receptor is a unique purinergic receptor that is exclusively expressed by microglia in the central nervous system (CNS). In this study, we set forth to investigate the role of P2Y12 receptors in microglial electrophysiological and morphological (static and dynamic) activation during spinal nerve transection (SNT)-induced neuropathic pain in mice. First, we found that a genetic deficiency of the P2Y12 receptor (P2Y12(-/-) mice) ameliorated pain hypersensitivities during the initiation phase of neuropathic pain. Next, we characterised both the electrophysiological and morphological properties of microglia in the superficial spinal cord dorsal horn following SNT injury. We show dramatic alterations including a peak at 3days post injury in microglial electrophysiology while high resolution two-photon imaging revealed significant changes of both static and dynamic microglial morphological properties by 7days post injury. Finally, in P2Y12(-/-) mice, these electrophysiological and morphological changes were ameliorated suggesting roles for P2Y12 receptors in SNT-induced microglial activation. Our results therefore indicate that P2Y12 receptors regulate microglial electrophysiological as well as static and dynamic microglial properties after peripheral nerve injury, suggesting that the microglial P2Y12 receptor could be a potential therapeutic target for the treatment of neuropathic pain.
Collapse
|
26
|
Eyo UB, Miner SA, Weiner JA, Dailey ME. Developmental changes in microglial mobilization are independent of apoptosis in the neonatal mouse hippocampus. Brain Behav Immun 2016; 55:49-59. [PMID: 26576723 PMCID: PMC4864211 DOI: 10.1016/j.bbi.2015.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/09/2015] [Accepted: 11/09/2015] [Indexed: 12/31/2022] Open
Abstract
During CNS development, microglia transform from highly mobile amoeboid-like cells to primitive ramified forms and, finally, to highly branched but relatively stationary cells in maturity. The factors that control developmental changes in microglia are largely unknown. Because microglia detect and clear apoptotic cells, developmental changes in microglia may be controlled by neuronal apoptosis. Here, we assessed the extent to which microglial cell density, morphology, motility, and migration are regulated by developmental apoptosis, focusing on the first postnatal week in the mouse hippocampus when the density of apoptotic bodies peaks at postnatal day 4 and declines sharply thereafter. Analysis of microglial form and distribution in situ over the first postnatal week showed that, although there was little change in the number of primary microglial branches, microglial cell density increased significantly, and microglia were often seen near or engulfing apoptotic bodies. Time-lapse imaging in hippocampal slices harvested at different times over the first postnatal week showed differences in microglial motility and migration that correlated with the density of apoptotic bodies. The extent to which these changes in microglia are driven by developmental neuronal apoptosis was assessed in tissues from BAX null mice lacking apoptosis. We found that apoptosis can lead to local microglial accumulation near apoptotic neurons in the pyramidal cell body layer but, unexpectedly, loss of apoptosis did not alter overall microglial cell density in vivo or microglial motility and migration in ex vivo tissue slices. These results demonstrate that developmental changes in microglial form, distribution, motility, and migration occur essentially normally in the absence of developmental apoptosis, indicating that factors other than neuronal apoptosis regulate these features of microglial development.
Collapse
|
27
|
The role of regulatory T cells and microglia in glioblastoma-associated immunosuppression. J Neurooncol 2015; 123:405-12. [PMID: 26123363 DOI: 10.1007/s11060-015-1849-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 06/08/2015] [Indexed: 01/09/2023]
Abstract
Cell-mediated suppression of anti-tumor immunity is multifactorial in patients with cancer, and recent studies have focused on several distinct cellular agents that are associated with this phenomenon. This review will focus on the potential role of regulatory T cells (Tregs) and microglia in the suppression of cellular immunity observed in patients with glioblastoma. We discuss the ontogeny, basic biology, evidence for activity, and potential clinical options for targeting Tregs and microglia as part of immunotherapy in affected patients.
Collapse
|
28
|
Madry C, Attwell D. Receptors, ion channels, and signaling mechanisms underlying microglial dynamics. J Biol Chem 2015; 290:12443-50. [PMID: 25855789 DOI: 10.1074/jbc.r115.637157] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microglia, the innate immune cells of the CNS, play a pivotal role in brain injury and disease. Microglia are extremely motile; their highly ramified processes constantly survey the brain parenchyma, and they respond promptly to brain damage with targeted process movement toward the injury site. Microglia play a key role in brain development and function by pruning synapses during development, phagocytosing apoptotic newborn neurons, and regulating neuronal activity by direct microglia-neuron or indirect microglia-astrocyte-neuron interactions, which all depend on their process motility. This review highlights recent discoveries about microglial dynamics, focusing on the receptors, ion channels, and signaling pathways involved.
Collapse
Affiliation(s)
- Christian Madry
- From the Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - David Attwell
- From the Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| |
Collapse
|
29
|
Reduced inflammatory phenotype in microglia derived from neonatal rat spinal cord versus brain. PLoS One 2014; 9:e99443. [PMID: 24914808 PMCID: PMC4051776 DOI: 10.1371/journal.pone.0099443] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 05/15/2014] [Indexed: 12/22/2022] Open
Abstract
Microglia are the primary immune cells of the central nervous system (CNS). Membrane bound sensors on their processes monitor the extracellular environment and respond to perturbations of the CNS such as injury or infection. Once activated, microglia play a crucial role in determining neuronal survival. Recent studies suggest that microglial functional response properties vary across different regions of the CNS. However, the activation profiles of microglia derived from the spinal cord have not been evaluated against brain microglia in vitro. Here, we studied the morphological properties and secretion of inflammatory and trophic effectors by microglia derived from the brain or spinal cord of neonatal rats under basal culture conditions and after activation with lipopolysaccharide (LPS). Our results demonstrate that spinal microglia assume a less inflammatory phenotype after LPS activation, with reduced release of the inflammatory effectors tumor necrosis factor alpha, interleukin-1 beta, and nitric oxide, a less amoeboid morphology, and reduced phagocytosis relative to brain-derived microglia. Phenotypic differences between brain and spinal microglia are an important consideration when evaluating anti-inflammatory or immunomodulatory therapies for brain versus spinal injury.
Collapse
|
30
|
Neher JJ, Neniskyte U, Hornik T, Brown GC. Inhibition of UDP/P2Y6 purinergic signaling prevents phagocytosis of viable neurons by activated microglia in vitro and in vivo. Glia 2014; 62:1463-75. [PMID: 24838858 PMCID: PMC4336556 DOI: 10.1002/glia.22693] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 04/30/2014] [Accepted: 05/02/2014] [Indexed: 11/25/2022]
Abstract
Microglia activated through Toll-like receptor (TLR)-2 or -4 can cause neuronal death by phagocytosing otherwise-viable neurons—a form of cell death called “phagoptosis.” UDP release from neurons has been shown to provoke microglial phagocytosis of neurons via microglial P2Y6 receptors, but whether inhibition of this process affects neuronal survival is unknown. We tested here whether inhibition of P2Y6 signaling could prevent neuronal death in inflammatory conditions, and whether UDP signaling can induce phagoptosis of stressed but viable neurons. We find that delayed neuronal loss and death in mixed neuronal/glial cultures induced by the TLR ligands lipopolysaccharide (LPS) or lipoteichoic acid was prevented by: apyrase (to degrade nucleotides), Reactive Blue 2 (to inhibit purinergic signaling), or MRS2578 (to specifically block P2Y6 receptors). In each case, inflammatory activation of microglia was not affected, and the rescued neurons remained viable for at least 7 days. Blocking P2Y6 receptors with MRS2578 also prevented phagoptosis of neurons induced by 250 nM amyloid beta 1–42, 5 μM peroxynitrite, or 50 μM 3-morpholinosydnonimine (which releases reactive oxygen and nitrogen species). Furthermore, the P2Y6 receptor agonist UDP by itself was sufficient to stimulate microglial phagocytosis and to induce rapid neuronal loss that was prevented by eliminating microglia or inhibiting phagocytosis. In vivo, injection of LPS into rat striatum induced microglial activation and delayed neuronal loss and blocking P2Y6 receptors with MRS2578 prevented this neuronal loss. Thus, blocking UDP/P2Y6 signaling is sufficient to prevent neuronal loss and death induced by a wide range of stimuli that activate microglial phagocytosis of neurons.
Collapse
Affiliation(s)
- Jonas J Neher
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
31
|
Ma Y, Cao W, Wang L, Jiang J, Nie H, Wang B, Wei X, Ying W. Basal CD38/cyclic ADP-ribose-dependent signaling mediates ATP release and survival of microglia by modulating connexin 43 hemichannels. Glia 2014; 62:943-55. [PMID: 24578339 DOI: 10.1002/glia.22651] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 02/04/2014] [Accepted: 02/05/2014] [Indexed: 02/06/2023]
Abstract
It is necessary to investigate the mechanisms underlying ATP release from neural cells, because extracellular ATP plays multiple important biological roles in the brain. CD38 is an ectoenzyme that consumes NAD(+) to produce cyclic ADP-ribose (cADPR), a potent agonist of ryanodine receptors. Our previous study showed that CD38 reductions led to microglial apoptosis. In this study, we used both murine microglial BV2 cells and primary microglial cultures as cellular models to test our hypothesis that basal CD38/cyclic ADP-ribose (CD38/cADPR)-dependent signaling plays a key role in ATP release, which mediates basal survival of microglia. We found that inhibition of CD38/cADPR-dependent signaling by CD38 silencing or 8-Bromo-cADPR, a ryanodine receptor antagonist, produced significant ATP release from BV2 microglia. Cx43 small interfering RNA and Cx43 hemichannel blocker 18-α-glycyrrhetinic acid completely prevented the CD38 silencing or 8-Bromo-cADPR-induced ATP release. Prevention of the ATP release could also be due to P2X7 receptor antagonists. Our study has further suggested a key role of ATP release in the microglial apoptosis induced by decreased CD38/cADPR-dependent signaling. In addition, by using primary microglial cultures, we found that 8-Bromo-cADPR also induced significant ATP release, which could be attenuated by 18-α-glycyrrhetinic acid. 8-Bromo-cADPR was also found to induce death of primary microglial cultures. In conclusion, our results have suggested novel roles of basal activation of CD38/cADPR-dependent signaling in mediating microglial functions and survival: It mediates ATP release from microglia by modulating Cx43 hemichannels, which can significantly affect microglial survival.
Collapse
Affiliation(s)
- Yingxin Ma
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Sugimoto K, Nishioka R, Ikeda A, Mise A, Takahashi H, Yano H, Kumon Y, Ohnishi T, Tanaka J. Activated microglia in a rat stroke model express NG2 proteoglycan in peri-infarct tissue through the involvement of TGF-β1. Glia 2013; 62:185-98. [PMID: 24311432 DOI: 10.1002/glia.22598] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/20/2013] [Accepted: 10/22/2013] [Indexed: 12/12/2022]
Abstract
We investigated activated microglia in ischemic brain lesions from rats that had been subjected to transient middle cerebral artery occlusion. Activated microglia expressing NG2 chondroitin sulfate proteoglycan (NG2) were found only in the narrow zone (demarcation zone) that demarcated the peri-infarct tissue and ischemic core. NG2(-) activated microglia were abundantly distributed in the peri-infarct tissue outside the demarcation zone. NG2(+) microglia but not NG2(-) microglia expressed both CD68 and a triggering receptor expressed on myeloid cells 2 (TREM-2), suggesting that NG2(+) microglia eliminated apoptotic neurons. In fact, NG2(+) microglia often attached to degenerating neurons and sometimes internalized NeuN(+) or neurofilament protein(+) material. Kinetic studies using quantitative real-time RT-PCR revealed that expression of transforming growth factor-β1 (TGF-β1) was most evident in the ischemic core; with this marker produced mainly by macrophages located in this region. TGF-β receptor mRNA expression peaked at 3 days post reperfusion (dpr) in the peri-infarct tissue, including the demarcation zone. Primary cultured rat microglia also expressed the receptor mRNA. In response to TGF-β1, primary microglia enhanced the expression of NG2 protein and TREM-2 mRNA as well as migratory activity. A TGF-β1 inhibitor, SB525334, abolished these effects. The present results suggest that TGF-β1 produced in the ischemic core diffused toward the peri-infarct tissue, driving activated microglial cells to eliminate degenerating neurons. Appropriate control of NG2(+) microglia in the demarcation zone might be a novel target for the suppression of secondary neurodegeneration in the peri-infarct tissue.
Collapse
Affiliation(s)
- Kana Sugimoto
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Dailey ME, Eyo U, Fuller L, Hass J, Kurpius D. Imaging microglia in brain slices and slice cultures. Cold Spring Harb Protoc 2013; 2013:1142-1148. [PMID: 24298036 DOI: 10.1101/pdb.prot079483] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Here we describe a method for imaging fluorescently labeled parenchymal microglia (MG) in excised neonatal or adult rodent brain tissue slices. Using multichannel confocal or two-photon time-lapse imaging, the approach affords real-time analyses of MG behaviors, including motility, migration, chemotaxis, proliferation, and phagocytosis in live brain tissues. The method is applicable to acutely prepared tissue slices from developing and adult rodents and to slice cultures derived from neonatal rodents, including transgenic and green fluorescent protein reporter mice. A variety of fluorescent tags can be used to study the structure and physiology of MG in these preparations. Moreover, bath application of reagents (such as ATP) can establish spatial and temporal gradients that induce chemokinesis- and chemotaxis-like MG migration in tissue slices. Thus, the approach is useful for dissecting the molecular basis of MG behaviors and testing whether candidate reagents alter MG behavior and function in semi-intact central nervous system tissue preparations.
Collapse
|
34
|
Bidirectional microglia-neuron communication in the healthy brain. Neural Plast 2013; 2013:456857. [PMID: 24078884 PMCID: PMC3775394 DOI: 10.1155/2013/456857] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 08/01/2013] [Indexed: 01/01/2023] Open
Abstract
Unlike other resident neural cells that are of neuroectodermal origin, microglia are resident neural cells of mesodermal origin. Traditionally recognized for their immune functions during disease, new roles are being attributed to these cells in the development and maintenance of the central nervous system (CNS) including specific communication with neurons. In this review, we highlight some of the recent findings on the bidirectional interaction between neurons and microglia. We discuss these interactions along two lines. First, we review data that suggest that microglial activity is modulated by neuronal signals, focusing on evidence that (i) neurons are capable of regulating microglial activation state and influence basal microglial activities; (ii) classic neurotransmitters affect microglial behavior; (iii) chemotactic signals attract microglia during acute neuronal injury. Next, we discuss some of the recent data on how microglia signal to neurons. Signaling mechanisms include (i) direct physical contact of microglial processes with neuronal elements; (ii) microglial regulation of neuronal synapse and circuit by fractalkine, complement, and DAP12 signaling. In addition, we discuss the use of microglial depletion strategies in studying the role of microglia in neuronal development and synaptic physiology. Deciphering the mechanisms of bidirectional microglial-neuronal communication provides novel insights in understanding microglial function in both the healthy and diseased brain.
Collapse
|
35
|
Morris GP, Clark IA, Zinn R, Vissel B. Microglia: a new frontier for synaptic plasticity, learning and memory, and neurodegenerative disease research. Neurobiol Learn Mem 2013; 105:40-53. [PMID: 23850597 DOI: 10.1016/j.nlm.2013.07.002] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 07/03/2013] [Indexed: 12/25/2022]
Abstract
We focus on emerging roles for microglia in synaptic plasticity, cognition and disease. We outline evidence that ramified microglia, traditionally thought to be functionally "resting" (i.e. quiescent) in the normal brain, in fact are highly dynamic and plastic. Ramified microglia continually and rapidly extend processes, contact synapses in an activity and experience dependent manner, and play a functionally dynamic role in synaptic plasticity, possibly through release of cytokines and growth factors. Ramified microglial also contribute to structural plasticity through the elimination of synapses via phagocytic mechanisms, which is necessary for normal cognition. Microglia have numerous mechanisms to monitor neuronal activity and numerous mechanisms also exist to prevent them transitioning to an activated state, which involves retraction of their surveying processes. Based on the evidence, we suggest that maintaining the ramified state of microglia is essential for normal synaptic and structural plasticity that supports cognition. Further, we propose that change of their ramified morphology and function, as occurs in inflammation associated with numerous neurological disorders such as Alzheimer's and Parkinson's disease, disrupts their intricate and essential synaptic functions. In turn altered microglia function could cause synaptic dysfunction and excess synapse loss early in disease, initiating a range of pathologies that follow. We conclude that the future of learning and memory research depends on an understanding of the role of non-neuronal cells and that this should include using sophisticated molecular, cellular, physiological and behavioural approaches combined with imaging to causally link the role of microglia to brain function and disease including Alzheimer's and Parkinson's disease and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gary P Morris
- Neurodegenerative Disorders, Garvan Institute of Medical Research, Neuroscience Department, Sydney, Australia; Faculty of Medicine, University of New South Wales, Sydney, Australia.
| | | | | | | |
Collapse
|
36
|
Li Z, Li W, Li Q, Tang M. Extracellular nucleotides and adenosine regulate microglial motility and their role in cerebral ischemia. Acta Pharm Sin B 2013. [DOI: 10.1016/j.apsb.2013.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
37
|
Ferreira R, Schlichter LC. Selective activation of KCa3.1 and CRAC channels by P2Y2 receptors promotes Ca(2+) signaling, store refilling and migration of rat microglial cells. PLoS One 2013; 8:e62345. [PMID: 23620825 PMCID: PMC3631179 DOI: 10.1371/journal.pone.0062345] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 03/20/2013] [Indexed: 12/26/2022] Open
Abstract
Microglial activation involves Ca(2+) signaling, and numerous receptors can evoke elevation of intracellular Ca(2+). ATP released from damaged brain cells can activate ionotropic and metabotropic purinergic receptors, and act as a chemoattractant for microglia. Metabotropic P2Y receptors evoke a Ca(2+) rise through release from intracellular Ca(2+) stores and store-operated Ca(2+) entry, and some have been implicated in microglial migration. This Ca(2+) rise is expected to activate small-conductance Ca(2+)-dependent K(+) (SK) channels, if present. We previously found that SK3 (KCa2.3) and KCa3.1 (SK4/IK1) are expressed in rat microglia and contribute to LPS-mediated activation and neurotoxicity. However, neither current has been studied by elevating Ca(2+) during whole-cell recordings. We hypothesized that, rather than responding only to Ca(2+), each channel type might be coupled to different receptor-mediated pathways. Here, our objective was to determine whether the channels are differentially activated by P2Y receptors, and, if so, whether they play differing roles. We used primary rat microglia and a rat microglial cell line (MLS-9) in which riluzole robustly activates both SK3 and KCa3.1 currents. Using electrophysiological, Ca(2+) imaging and pharmacological approaches, we show selective functional coupling of KCa3.1 to UTP-mediated P2Y2 receptor activation. KCa3.1 current is activated by Ca(2+) entry through Ca(2+)-release-activated Ca(2+) (CRAC/Orai1) channels, and both CRAC/Orai1 and KCa3.1 channels facilitate refilling of Ca(2+) stores. The Ca(2+) dependence of KCa3.1 channel activation was skewed to abnormally high concentrations, and we present evidence for a close physical association of the two channel types. Finally, migration of primary rat microglia was stimulated by UTP and inhibited by blocking either KCa3.1 or CRAC/Orai1 channels. This is the first report of selective coupling of one type of SK channel to purinergic stimulation of microglia, transactivation of KCa3.1 channels by CRAC/Orai1, and coordinated roles for both channels in store refilling, Ca(2+) signaling and microglial migration.
Collapse
Affiliation(s)
- Roger Ferreira
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Lyanne C. Schlichter
- Genes and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Eyo UB, Dailey ME. Microglia: key elements in neural development, plasticity, and pathology. J Neuroimmune Pharmacol 2013; 8:494-509. [PMID: 23354784 DOI: 10.1007/s11481-013-9434-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 01/14/2013] [Indexed: 12/31/2022]
Abstract
A century after Cajal identified a "third element" of the nervous system, many issues have been clarified about the identity and function of one of its major components, the microglia. Here, we review recent findings by microgliologists, highlighting results from imaging studies that are helping provide new views of microglial behavior and function. In vivo imaging in the intact adult rodent CNS has revolutionized our understanding of microglial behaviors in situ and has raised speculation about their function in the uninjured adult brain. Imaging studies in ex vivo mammalian tissue preparations and in intact model organisms including zebrafish are providing insights into microglial behaviors during brain development. These data suggest that microglia play important developmental roles in synapse remodeling, developmental apoptosis, phagocytic clearance, and angiogenesis. Because microglia also contribute to pathology, including neurodevelopmental and neurobehavioral disorders, ischemic injury, and neuropathic pain, promising new results raise the possibility of leveraging microglia for therapeutic roles. Finally, exciting recent work is addressing unanswered questions regarding the nature of microglial-neuronal communication. While it is now apparent that microglia play diverse roles in neural development, behavior, and pathology, future research using neuroimaging techniques will be essential to more fully exploit these intriguing cellular targets for effective therapeutic intervention applied to a variety of conditions.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
39
|
Morrison HW, Filosa JA. A quantitative spatiotemporal analysis of microglia morphology during ischemic stroke and reperfusion. J Neuroinflammation 2013; 10:4. [PMID: 23311642 PMCID: PMC3570327 DOI: 10.1186/1742-2094-10-4] [Citation(s) in RCA: 381] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 12/24/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Microglia cells continuously survey the healthy brain in a ramified morphology and, in response to injury, undergo progressive morphological and functional changes that encompass microglia activation. Although ideally positioned for immediate response to ischemic stroke (IS) and reperfusion, their progressive morphological transformation into activated cells has not been quantified. In addition, it is not well understood if diverse microglia morphologies correlate to diverse microglia functions. As such, the dichotomous nature of these cells continues to confound our understanding of microglia-mediated injury after IS and reperfusion. The purpose of this study was to quantitatively characterize the spatiotemporal pattern of microglia morphology during the evolution of cerebral injury after IS and reperfusion. METHODS Male C57Bl/6 mice were subjected to focal cerebral ischemia and periods of reperfusion (0, 8 and 24 h). The microglia process length/cell and number of endpoints/cell was quantified from immunofluorescent confocal images of brain regions using a skeleton analysis method developed for this study. Live cell morphology and process activity were measured from movies acquired in acute brain slices from GFP-CX3CR1 transgenic mice after IS and 24-h reperfusion. Regional CD11b and iNOS expressions were measured from confocal images and Western blot, respectively, to assess microglia proinflammatory function. RESULTS Quantitative analysis reveals a significant spatiotemporal relationship between microglia morphology and evolving cerebral injury in the ipsilateral hemisphere after IS and reperfusion. Microglia were both hyper- and de-ramified in striatal and cortical brain regions (respectively) after 60 min of focal cerebral ischemia. However, a de-ramified morphology was prominent when ischemia was coupled to reperfusion. Live microglia were de-ramified, and, in addition, process activity was severely blunted proximal to the necrotic core after IS and 24 h of reperfusion. CD11b expression, but not iNOS expression, was increased in regions of hyper- and de-ramified microglia during the course of ischemic stroke and 24 h of reperfusion. CONCLUSIONS Our findings illustrate that microglia activation after stroke includes both increased and decreased cell ramification. Importantly, quantitative analyses of microglial morphology and activity are feasible and, in future studies, would assist in the comprehensive identification and stratification of their dichotomous contribution toward cerebral injury and recovery during IS and reperfusion.
Collapse
|
40
|
Su X, Floyd DH, Hughes A, Xiang J, Schneider JG, Uluckan O, Heller E, Deng H, Zou W, Craft CS, Wu K, Hirbe AC, Grabowska D, Eagleton MC, Townsley S, Collins L, Piwnica-Worms D, Steinberg TH, Novack DV, Conley PB, Hurchla MA, Rogers M, Weilbaecher KN. The ADP receptor P2RY12 regulates osteoclast function and pathologic bone remodeling. J Clin Invest 2012; 122:3579-92. [PMID: 22996695 DOI: 10.1172/jci38576] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 07/26/2012] [Indexed: 12/16/2022] Open
Abstract
The adenosine diphosphate (ADP) receptor P2RY12 (purinergic receptor P2Y, G protein coupled, 12) plays a critical role in platelet aggregation, and P2RY12 inhibitors are used clinically to prevent cardiac and cerebral thrombotic events. Extracellular ADP has also been shown to increase osteoclast (OC) activity, but the role of P2RY12 in OC biology is unknown. Here, we examined the role of mouse P2RY12 in OC function. Mice lacking P2ry12 had decreased OC activity and were partially protected from age-associated bone loss. P2ry12-/- OCs exhibited intact differentiation markers, but diminished resorptive function. Extracellular ADP enhanced OC adhesion and resorptive activity of WT, but not P2ry12-/-, OCs. In platelets, ADP stimulation of P2RY12 resulted in GTPase Ras-related protein (RAP1) activation and subsequent αIIbβ3 integrin activation. Likewise, we found that ADP stimulation induced RAP1 activation in WT and integrin β3 gene knockout (Itgb3-/-) OCs, but its effects were substantially blunted in P2ry12-/- OCs. In vivo, P2ry12-/- mice were partially protected from pathologic bone loss associated with serum transfer arthritis, tumor growth in bone, and ovariectomy-induced osteoporosis: all conditions associated with increased extracellular ADP. Finally, mice treated with the clinical inhibitor of P2RY12, clopidogrel, were protected from pathologic osteolysis. These results demonstrate that P2RY12 is the primary ADP receptor in OCs and suggest that P2RY12 inhibition is a potential therapeutic target for pathologic bone loss.
Collapse
Affiliation(s)
- Xinming Su
- Department of Medicine, Division of Oncology, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Corriden R, Insel PA. New insights regarding the regulation of chemotaxis by nucleotides, adenosine, and their receptors. Purinergic Signal 2012; 8:587-98. [PMID: 22528684 PMCID: PMC3360098 DOI: 10.1007/s11302-012-9311-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 01/05/2012] [Indexed: 12/23/2022] Open
Abstract
The directional movement of cells can be regulated by ATP, certain other nucleotides (e.g., ADP, UTP), and adenosine. Such regulation occurs for cells that are "professional phagocytes" (e.g., neutrophils, macrophages, certain lymphocytes, and microglia) and that undergo directional migration and subsequent phagocytosis. Numerous other cell types (e.g., fibroblasts, endothelial cells, neurons, and keratinocytes) also change motility and migration in response to ATP, other nucleotides, and adenosine. In this article, we review how nucleotides and adenosine modulate chemotaxis and motility and highlight the importance of nucleotide- and adenosine-regulated cell migration in several cell types: neutrophils, microglia, endothelial cells, and cancer cells. We also discuss difficulties in conducting experiments and drawing conclusions regarding the ability of nucleotides and adenosine to modulate the migration of professional and non-professional phagocytes.
Collapse
Affiliation(s)
- Ross Corriden
- Institute of Cell Signalling, University of Nottingham, Nottingham, UK
| | - Paul A. Insel
- Departments of Pharmacology and Medicine, University of California, San Diego, CA USA
- Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, Mail code 0636, La Jolla, CA 92093 USA
| |
Collapse
|
42
|
Eyo U, Dailey ME. Effects of oxygen-glucose deprivation on microglial mobility and viability in developing mouse hippocampal tissues. Glia 2012; 60:1747-60. [PMID: 22847985 DOI: 10.1002/glia.22394] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 06/27/2012] [Indexed: 12/29/2022]
Abstract
As brain-resident immune cells, microglia (MG) survey the brain parenchyma to maintain homeostasis during development and following injury. Research in perinatal stroke, a leading cause of lifelong disability, has implicated MG as targets for therapeutic intervention during stroke. Although MG responses are complex, work in developing rodents suggests that MG limit brain damage after stroke. However, little is known about how energy-limiting conditions affect MG survival and mobility (motility and migration) in developing brain tissues. Here, we used confocal time-lapse imaging to monitor MG viability and mobility during hypoxia or oxygen-glucose deprivation (OGD) in hippocampal tissue slices derived from neonatal GFP-reporter mice (CX3CR1(GFP/+) ). We found that MG remain viable for at least 6 h of hypoxia but begin to die after 2 h of OGD, while both hypoxia and OGD reduce MG motility. Unexpectedly, some MG retain or recover motility during OGD and can engulf dead cells. Additionally, MG from younger neonates (P2-P3) are more resistant to OGD than those from older ones (P6-P7), indicating increasing vulnerability with developmental age. Finally, transient (2 h) OGD also increases MG death, and although motility is rapidly restored after transient OGD, it remains below control levels for many hours. Together, these results show that MG in neonatal mouse brain tissues are vulnerable to both transient and sustained OGD, and many MG die within hours after onset of OGD. Preventing MG death may, therefore, provide a strategy for promoting tissue restoration after stroke.
Collapse
Affiliation(s)
- Ukpong Eyo
- Department of Biology, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
43
|
Brites D. The evolving landscape of neurotoxicity by unconjugated bilirubin: role of glial cells and inflammation. Front Pharmacol 2012; 3:88. [PMID: 22661946 PMCID: PMC3361682 DOI: 10.3389/fphar.2012.00088] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 04/23/2012] [Indexed: 12/13/2022] Open
Abstract
Unconjugated hyperbilirubinemia is a common condition in the first week of postnatal life. Although generally harmless, some neonates may develop very high levels of unconjugated bilirubin (UCB), which may surpass the protective mechanisms of the brain in preventing UCB accumulation. In this case, both short-term and long-term neurodevelopmental disabilities, such as acute and chronic UCB encephalopathy, known as kernicterus, or more subtle alterations defined as bilirubin-induced neurological dysfunction (BIND) may be produced. There is a tremendous variability in babies' vulnerability toward UCB for reasons not yet explained, but preterm birth, sepsis, hypoxia, and hemolytic disease are comprised as risk factors. Therefore, UCB levels and neurological abnormalities are not strictly correlated. Even nowadays, the mechanisms of UCB neurotoxicity are still unclear, as are specific biomarkers, and little is known about lasting sequelae attributable to hyperbilirubinemia. On autopsy, UCB was shown to be within neurons, neuronal processes, and microglia, and to produce loss of neurons, demyelination, and gliosis. In isolated cell cultures, UCB was shown to impair neuronal arborization and to induce the release of pro-inflammatory cytokines from microglia and astrocytes. However, cell dependent sensitivity to UCB toxicity and the role of each nerve cell type remains not fully understood. This review provides a comprehensive insight into cell susceptibilities and molecular targets of UCB in neurons, astrocytes, and oligodendrocytes, and on phenotypic and functional responses of microglia to UCB. Interplay among glia elements and cross-talk with neurons, with a special emphasis in the UCB-induced immunostimulation, and the role of sepsis in BIND pathogenesis are highlighted. New and interesting data on the anti-inflammatory and antioxidant activities of different pharmacological agents are also presented, as novel and promising additional therapeutic approaches to BIND.
Collapse
Affiliation(s)
- Dora Brites
- Neuron Glia Biology in Health and Disease Unit, Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon Lisbon, Portugal
| |
Collapse
|
44
|
P2X4 purinoceptor signaling in chronic pain. Purinergic Signal 2012; 8:621-8. [PMID: 22528681 DOI: 10.1007/s11302-012-9306-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 02/14/2012] [Indexed: 01/19/2023] Open
Abstract
ATP, acting via P2 purinergic receptors, is a known mediator of inflammatory and neuropathic pain. There is increasing evidence that the ATP-gated P2X4 receptor (P2X4R) subtype is a locus through which activity of spinal microglia and peripheral macrophages instigate pain hypersensitivity caused by inflammation or by injury to a peripheral nerve. The present article highlights the recent advances in our understanding of microglia-neuron interactions in neuropathic pain by focusing on the signaling and regulation of the P2X4R. We will also develop a framework for understanding converging lines of evidence for involvement of P2X4Rs expressed on macrophages in peripheral inflammatory pain.
Collapse
|
45
|
Ohsawa K, Sanagi T, Nakamura Y, Suzuki E, Inoue K, Kohsaka S. Adenosine A3 receptor is involved in ADP-induced microglial process extension and migration. J Neurochem 2012; 121:217-27. [PMID: 22335470 DOI: 10.1111/j.1471-4159.2012.07693.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The extension of microglial processes toward injured sites in the brain is triggered by the stimulation of the purinergic receptor P2Y(12) by extracellular ATP. We recently showed that P2Y(12) stimulation by ATP induces microglial process extension in collagen gels. In the present study, we found that a P2Y(12) agonist, 2-methylthio-ADP (2MeSADP), failed to induce the process extension of microglia in collagen gels and that co-stimulation with adenosine, a phosphohydrolytic derivative of ATP, and 2MeSADP restored the chemotactic process extension. An adenosine A3 receptor (A3R)-selective agonist restored the chemotactic process extension, but other receptor subtype agonists did not. The removal of adenosine by adenosine deaminase and the blocking of A3R by an A3R-selective antagonist inhibited ADP-induced process extension. The A3R antagonist inhibited ADP-induced microglial migration, and an A3R agonist promoted 2MeSADP-stimulated migration. ADP and the A3R agonist activated Jun N-terminal kinase in microglia, and a Jun N-terminal kinase inhibitor inhibited the ADP-induced process extension. An RT-PCR analysis showed that A1R and A3R were expressed by microglia sorted from adult rat brains and that the A2AR expression level was very low. These results suggested that A3R signaling may be involved in the ADP-induced process extension and migration of microglia.
Collapse
Affiliation(s)
- Keiko Ohsawa
- Department of Neurochemistry, National Institute of Neuroscience, 4-1-1 Ogawahigashi Kodaira, Tokyo 187-8502, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Carrasco MC, Navascués J, Cuadros MA, Calvente R, Martín-Oliva D, Santos AM, Sierra A, Ferrer-Martín RM, Marín-Teva JL. Migration and ramification of microglia in quail embryo retina organotypic cultures. Dev Neurobiol 2012; 71:296-315. [PMID: 21394933 DOI: 10.1002/dneu.20860] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Organotypic cultures of retina explants preserve the complex cellular microenvironment of the retina and have been used as a tool to assess the biological functions of some cell types. However, studies to date have shown that microglial cells activate quickly in response to the retina explantation. In this study, microglial cells migrated and ramified in quail embryo retina organotypic cultures (QEROCs) according to chronological patterns bearing a resemblance to those in the retina in situ, despite some differences in cell density and ramification degree. Retinal explants from quail embryos at 9 days of incubation (E9) proved to be the best in vitro system for reproducing a physiological-like behavior of microglial cells when cultured in Eagle's basal medium supplemented with horse serum. During the first week in vitro, microglial cells migrated tangentially in the vitreal part of QEROCs, and some began to migrate radially from 3 days in vitro (div) onward, ramifying in the inner and outer plexiform layers, thus mimicking microglia development in the retina in situ, although reaching a lower degree of ramification after 7 div. From 8 div onward, microglial cells rounded throughout the explant thickness simultaneously with the nonphysiological appearance of dead photoreceptors and round microglia in the outernuclear layer. Therefore, E9 QEROCs can be used during the first week in vitro as a model system for experimental studies of molecules putatively involved in microglial migration and ramification.
Collapse
Affiliation(s)
- Maria-Carmen Carrasco
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, E-18071 Granada, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Inoue K, Tsuda M. Purinergic systems, neuropathic pain and the role of microglia. Exp Neurol 2011; 234:293-301. [PMID: 21946271 DOI: 10.1016/j.expneurol.2011.09.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 09/01/2011] [Accepted: 09/09/2011] [Indexed: 01/23/2023]
Abstract
We have learned various data on the role of purinoceptors (P2X4, P2X7, P2Y6 and P2Y12) expressed in spinal microglia and several factors that presumably activate microglia in neuropathic pain after peripheral nerve injury. Purinergic receptor-mediated spinal microglial functions make a critical contribution to pathologically enhanced pain processing in the dorsal horn. Microglial purinoceptors might be promising targets for treating neuropathic pain. A predicted therapeutic benefit of interfering with microglial purinergic receptors may be that normal pain sensitivity would be unaffected since expression or activity of most of these receptors are upregulated or enhanced predominantly in activated microglia in the spinal cord where damaged sensory fibers project.
Collapse
Affiliation(s)
- Kazuhide Inoue
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi, Fukuoka 812-8582, Japan.
| | | |
Collapse
|
48
|
Masuda T, Croom D, Hida H, Kirov SA. Capillary blood flow around microglial somata determines dynamics of microglial processes in ischemic conditions. Glia 2011; 59:1744-53. [PMID: 21800362 DOI: 10.1002/glia.21220] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 06/21/2011] [Indexed: 11/06/2022]
Abstract
Microglia are the resident immune cells in the brain. Under normal conditions, resting ramified microglia constantly extend and retract fine processes while performing immunological surveillance. In ischemia, microglia become activated as demonstrated by morphological changes during deramification leading to transformation from ramified to amoeboid form. In vivo two-photon microscopy of enhanced green fluorescent protein (EGFP)-expressing microglia in mouse neocortex was used to examine microglial dynamics during the early periods of focal and global ischemia. A penumbra-like "area-at-risk" surrounded by a square-shaped area of severely hypoperfused tissue was created by laser-induced photothrombosis. The dynamics of microglial processes in the area-at-risk was strongly correlated with capillary blood flow (BF) measured within 10 μm of microglial somata. Changes in BF around distal microglial processes (>30 μm from somata) had no effect on microglial dynamics. A severe reduction of capillary BF near somata by 84% ± 6% resulted in initiation of microglial deramification, suggesting activation. A moderate decrease in BF near somata by 22% ± 5% or increase by 87% ± 10%, reflecting a redistribution of capillary BF, had no effect on microglial morphology. Complete BF loss during cardiac arrest (CA) or transient bilateral common carotid artery occlusion (BCCAO) entirely stalled all microglial processes without structural changes. Reperfusion after BCCAO induced recovery of microglial dynamics to preocclusion values. These findings suggest that during ischemia, the severe drop in BF around microglial somata coincides with morphological activation. However, this activation requires some residual BF, because complete perfusion loss (as during BCCAO and CA) did not support microglial deramification.
Collapse
Affiliation(s)
- Tadashi Masuda
- Brain and Behavior Discovery Institute, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | |
Collapse
|
49
|
van Weering HRJ, Boddeke HWGM, Vinet J, Brouwer N, de Haas AH, van Rooijen N, Thomsen AR, Biber KPH. CXCL10/CXCR3 signaling in glia cells differentially affects NMDA-induced cell death in CA and DG neurons of the mouse hippocampus. Hippocampus 2011; 21:220-32. [PMID: 20082289 DOI: 10.1002/hipo.20742] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The chemokine CXCL10 and its receptor CXCR3 are implicated in various CNS pathologies since interference with CXCL10/CXCR3 signaling alters the onset and progression in various CNS disease models. However, the mechanism and cell-types involved in CXCL10/CXCR3 signaling under pathological conditions are far from understood. Here, we investigated the potential role for CXCL10/CXCR3 signaling in neuronal cell death and glia activation in response to N-methyl-D-aspartic acid (NMDA)-induced excitotoxicity in mouse organotypic hippocampal slice cultures (OHSCs). Our findings demonstrate that astrocytes express CXCL10 in response to excitotoxicity. Experiments in OHSCs derived from CXCL10-deficient (CXCL10(-/-) ) and CXCR3-deficient (CXCR3(-/-) ) revealed that in the absence of CXCL10 or CXCR3, neuronal cell death in the CA1 and CA3 regions was diminished after NMDA-treatment when compared to wild type OHSCs. In contrast, neuronal cell death in the DG region was enhanced in both CXCL10(-/-) and CXCR3(-/-) OHSCs in response to a high (50 μM) NMDA-concentration. Moreover, we show that in the absence of microglia the differential changes in neuronal vulnerability between CXCR3(-/-) and wild type OHSCs are fully abrogated and therefore a prominent role for microglia in this process is suggested. Taken together, our results identify a region-specific role for CXCL10/CXCR3 signaling in neuron-glia and glia-glia interactions under pathological conditions.
Collapse
Affiliation(s)
- Hilmar R J van Weering
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen (UMCG), Rijksuniversiteit Groningen (RUG), Groningen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kraft AD, Kaltenbach LS, Lo DC, Harry GJ. Activated microglia proliferate at neurites of mutant huntingtin-expressing neurons. Neurobiol Aging 2011; 33:621.e17-33. [PMID: 21482444 DOI: 10.1016/j.neurobiolaging.2011.02.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 02/01/2011] [Accepted: 02/16/2011] [Indexed: 01/13/2023]
Abstract
In Huntington's disease (HD), mutated huntingtin (mhtt) causes striatal neurodegeneration which is paralleled by elevated microglia cell numbers. In vitro corticostriatal slice and primary neuronal culture models, in which neuronal expression of mhtt fragments drives HD-like neurotoxicity, were employed to examine wild type microglia during both the initiation and progression of neuronal pathology. As neuronal pathology progressed, microglia initially localized in the vicinity of neurons expressing mhtt fragments increased in number, demonstrated morphological evidence of activation, and expressed the proliferation marker, Ki67. These microglia were positioned along irregular neurites, but did not localize with mhtt inclusions nor exacerbate mhtt fragment-induced neurotoxicity. Prior to neuronal pathology, microglia upregulated ionized calcium binding adaptor molecule 1 (Iba1), signaling a functional shift. With neurodegeneration, interleukin-6 and complement component 1q were increased. The results suggest a stimulatory, proliferative signal for microglia present at the onset of mhtt fragment-induced neurodegeneration. Thus, microglia effect a localized inflammatory response to neuronal mhtt expression that may serve to direct microglial removal of dysfunctional neurites or aberrant synapses, as is required for reparative actions in vivo.
Collapse
Affiliation(s)
- Andrew D Kraft
- Neurotoxicology Group, Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | | | | | |
Collapse
|