351
|
Wohleb ES, Franklin T, Iwata M, Duman RS. Integrating neuroimmune systems in the neurobiology of depression. Nat Rev Neurosci 2016; 17:497-511. [PMID: 27277867 DOI: 10.1038/nrn.2016.69] [Citation(s) in RCA: 453] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Data from clinical and preclinical studies indicate that immune dysregulation, specifically of inflammatory processes, is associated with symptoms of major depressive disorder (MDD). In particular, increased levels of circulating pro-inflammatory cytokines and concomitant activation of brain-resident microglia can lead to depressive behavioural symptoms. Repeated exposure to psychological stress has a profound impact on peripheral immune responses and perturbs the function of brain microglia, which may contribute to neurobiological changes underlying MDD. Here, we review these findings and discuss ongoing studies examining neuroimmune mechanisms that influence neuronal activity as well as synaptic plasticity. Interventions targeting immune-related cellular and molecular pathways may benefit subsets of MDD patients with immune dysregulation.
Collapse
Affiliation(s)
- Eric S Wohleb
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, Connecticut 06519, USA
| | - Tina Franklin
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, Connecticut 06519, USA
| | - Masaaki Iwata
- Division of Neuropsychiatry, Department of Brain and Neurosciences, Tottori University Faculty of Medicine, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Ronald S Duman
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, Connecticut 06519, USA
| |
Collapse
|
352
|
Tay TL, Savage JC, Hui CW, Bisht K, Tremblay MÈ. Microglia across the lifespan: from origin to function in brain development, plasticity and cognition. J Physiol 2016; 595:1929-1945. [PMID: 27104646 DOI: 10.1113/jp272134] [Citation(s) in RCA: 393] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/15/2016] [Indexed: 12/11/2022] Open
Abstract
Microglia are the only immune cells that permanently reside in the central nervous system (CNS) alongside neurons and other types of glial cells. The past decade has witnessed a revolution in our understanding of their roles during normal physiological conditions. Cutting-edge techniques revealed that these resident immune cells are critical for proper brain development, actively maintain health in the mature brain, and rapidly adapt their function to physiological or pathophysiological needs. In this review, we highlight recent studies on microglial origin (from the embryonic yolk sac) and the factors regulating their differentiation and homeostasis upon brain invasion. Elegant experiments tracking microglia in the CNS allowed studies of their unique roles compared with other types of resident macrophages. Here we review the emerging roles of microglia in brain development, plasticity and cognition, and discuss the implications of the depletion or dysfunction of microglia for our understanding of disease pathogenesis. Immune activation, inflammation and various other conditions resulting in undesirable microglial activity at different stages of life could severely impair learning, memory and other essential cognitive functions. The diversity of microglial phenotypes across the lifespan, between compartments of the CNS, and sexes, as well as their crosstalk with the body and external environment, is also emphasised. Understanding what defines particular microglial phenotypes is of major importance for future development of innovative therapies controlling their effector functions, with consequences for cognition across chronic stress, ageing, neuropsychiatric and neurological diseases.
Collapse
Affiliation(s)
- Tuan Leng Tay
- Institute of Neuropathology, University of Freiburg, Germany
| | - Julie C Savage
- Axe Neurosciences, Centre de recherche du CHU de Québec, Québec, Canada
| | - Chin Wai Hui
- Axe Neurosciences, Centre de recherche du CHU de Québec, Québec, Canada
| | - Kanchan Bisht
- Axe Neurosciences, Centre de recherche du CHU de Québec, Québec, Canada
| | | |
Collapse
|
353
|
Eyo UB, Murugan M, Wu LJ. Microglia-Neuron Communication in Epilepsy. Glia 2016; 65:5-18. [PMID: 27189853 DOI: 10.1002/glia.23006] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/20/2016] [Accepted: 04/28/2016] [Indexed: 12/22/2022]
Abstract
Epilepsy has remained a significant social concern and financial burden globally. Current therapeutic strategies are based primarily on neurocentric mechanisms that have not proven successful in at least a third of patients, raising the need for novel alternative and complementary approaches. Recent evidence implicates glial cells and neuroinflammation in the pathogenesis of epilepsy with the promise of targeting these cells to complement existing strategies. Specifically, microglial involvement, as a major inflammatory cell in the epileptic brain, has been poorly studied. In this review, we highlight microglial reaction to experimental seizures, discuss microglial control of neuronal activities, and propose the functions of microglia during acute epileptic phenotypes, delayed neurodegeneration, and aberrant neurogenesis. Future research that would help fill in the current gaps in our knowledge includes epilepsy-induced alterations in basic microglial functions, neuro-microglial interactions during chronic epilepsy, and microglial contribution to developmental seizures. Studying the role of microglia in epilepsy could inform therapies to better alleviate the disease. GLIA 2016;65:5-18.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Madhuvika Murugan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Long-Jun Wu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
354
|
Szalay G, Martinecz B, Lénárt N, Környei Z, Orsolits B, Judák L, Császár E, Fekete R, West BL, Katona G, Rózsa B, Dénes Á. Microglia protect against brain injury and their selective elimination dysregulates neuronal network activity after stroke. Nat Commun 2016; 7:11499. [PMID: 27139776 PMCID: PMC4857403 DOI: 10.1038/ncomms11499] [Citation(s) in RCA: 463] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 04/04/2016] [Indexed: 12/15/2022] Open
Abstract
Microglia are the main immune cells of the brain and contribute to common brain diseases. However, it is unclear how microglia influence neuronal activity and survival in the injured brain in vivo. Here we develop a precisely controlled model of brain injury induced by cerebral ischaemia combined with fast in vivo two-photon calcium imaging and selective microglial manipulation. We show that selective elimination of microglia leads to a striking, 60% increase in infarct size, which is reversed by microglial repopulation. Microglia-mediated protection includes reduction of excitotoxic injury, since an absence of microglia leads to dysregulated neuronal calcium responses, calcium overload and increased neuronal death. Furthermore, the incidence of spreading depolarization (SD) is markedly reduced in the absence of microglia. Thus, microglia are involved in changes in neuronal network activity and SD after brain injury in vivo that could have important implications for common brain diseases.
Collapse
Affiliation(s)
- Gergely Szalay
- Two-Photon Imaging Center, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary
| | - Bernadett Martinecz
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary
| | - Nikolett Lénárt
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary
| | - Zsuzsanna Környei
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary
| | - Barbara Orsolits
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary
| | - Linda Judák
- Two-Photon Imaging Center, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary.,MTA-PPKE ITK-NAP B - Two-photon measurement Technology Research Group, Pázmány Péter University, Budapest 1083, Hungary
| | - Eszter Császár
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary
| | - Rebeka Fekete
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary
| | - Brian L West
- Plexxikon, Inc., Berkeley, California 94710, USA
| | - Gergely Katona
- MTA-PPKE ITK-NAP B - Two-photon measurement Technology Research Group, Pázmány Péter University, Budapest 1083, Hungary
| | - Balázs Rózsa
- Two-Photon Imaging Center, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary.,MTA-PPKE ITK-NAP B - Two-photon measurement Technology Research Group, Pázmány Péter University, Budapest 1083, Hungary
| | - Ádám Dénes
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony U. 43, Budapest 1083, Hungary
| |
Collapse
|
355
|
Liu C, Wu C, Yang Q, Gao J, Li L, Yang D, Luo L. Macrophages Mediate the Repair of Brain Vascular Rupture through Direct Physical Adhesion and Mechanical Traction. Immunity 2016; 44:1162-76. [PMID: 27156384 DOI: 10.1016/j.immuni.2016.03.008] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 01/11/2016] [Accepted: 03/22/2016] [Indexed: 01/11/2023]
Abstract
Hemorrhagic stroke and brain microbleeds are caused by cerebrovascular ruptures. Fast repair of such ruptures is the most promising therapeutic approach. Due to a lack of high-resolution in vivo real-time studies, the dynamic cellular events involved in cerebrovascular repair remain unknown. Here, we have developed a cerebrovascular rupture system in zebrafish by using multi-photon laser, which generates a lesion with two endothelial ends. In vivo time-lapse imaging showed that a macrophage arrived at the lesion and extended filopodia or lamellipodia to physically adhere to both endothelial ends. This macrophage generated mechanical traction forces to pull the endothelial ends and facilitate their ligation, thus mediating the repair of the rupture. Both depolymerization of microfilaments and inhibition of phosphatidylinositide 3-kinase or Rac1 activity disrupted macrophage-endothelial adhesion and impaired cerebrovascular repair. Our study reveals a hitherto unexpected role for macrophages in mediating repair of cerebrovascular ruptures through direct physical adhesion and mechanical traction.
Collapse
Affiliation(s)
- Chi Liu
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | - Chuan Wu
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | - Qifen Yang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | - Jing Gao
- Department of Endodontics and Operative Dentistry, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology, Chongqing Medical University, 401147 Chongqing, China
| | - Li Li
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | - Deqin Yang
- Department of Endodontics and Operative Dentistry, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, The Affiliated Hospital of Stomatology, Chongqing Medical University, 401147 Chongqing, China.
| | - Lingfei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China.
| |
Collapse
|
356
|
Tay TL, Hagemeyer N, Prinz M. The force awakens: insights into the origin and formation of microglia. Curr Opin Neurobiol 2016; 39:30-7. [PMID: 27107946 DOI: 10.1016/j.conb.2016.04.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 04/05/2016] [Accepted: 04/06/2016] [Indexed: 12/12/2022]
Abstract
Microglia are tissue resident macrophages of the central nervous system (CNS) that maintain homeostasis and respond to immune challenges. New genetic fate mapping tools have revealed a yolk sac origin of microglia. Once established in the CNS, microglia persist throughout the lifetime of the organism behind the blood-brain barrier and maintain themselves by self-renewal. Recent studies uncovered a broad spectrum of microglial functions that are influenced by the dynamism of brain formation and neuronal wiring. This review focuses on current findings concerning microglia origin and formation during development and discusses the factors important for microglia survival and function.
Collapse
Affiliation(s)
- Tuan Leng Tay
- Institute of Neuropathology, University of Freiburg, Germany
| | - Nora Hagemeyer
- Institute of Neuropathology, University of Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Germany.
| |
Collapse
|
357
|
Wu Y, Dissing-Olesen L, MacVicar BA, Stevens B. Microglia: Dynamic Mediators of Synapse Development and Plasticity. Trends Immunol 2016; 36:605-613. [PMID: 26431938 DOI: 10.1016/j.it.2015.08.008] [Citation(s) in RCA: 511] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/13/2015] [Accepted: 08/13/2015] [Indexed: 12/25/2022]
Abstract
Neuronal communication underlies all brain activity and the genesis of complex behavior. Emerging research has revealed an unexpected role for immune molecules in the development and plasticity of neuronal synapses. Moreover microglia, the resident immune cells of the brain, express and secrete immune-related signaling molecules that alter synaptic transmission and plasticity in the absence of inflammation. When inflammation does occur, microglia modify synaptic connections and synaptic plasticity required for learning and memory. Here we review recent findings demonstrating how the dynamic interactions between neurons and microglia shape the circuitry of the nervous system in the healthy brain and how altered neuron-microglia signaling could contribute to disease.
Collapse
Affiliation(s)
- Yuwen Wu
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA; These authors contributed equally to this work
| | - Lasse Dissing-Olesen
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 2B5, Canada; These authors contributed equally to this work
| | - Brian A MacVicar
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Beth Stevens
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
358
|
Frost JL, Schafer DP. Microglia: Architects of the Developing Nervous System. Trends Cell Biol 2016; 26:587-597. [PMID: 27004698 DOI: 10.1016/j.tcb.2016.02.006] [Citation(s) in RCA: 261] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/16/2016] [Accepted: 02/19/2016] [Indexed: 11/18/2022]
Abstract
Microglia are resident macrophages of the central nervous system (CNS), representing 5-10% of total CNS cells. Recent findings reveal that microglia enter the embryonic brain, take up residence before the differentiation of other CNS cell types, and become critical regulators of CNS development. Here, we discuss exciting new work implicating microglia in a range of developmental processes, including regulation of cell number and spatial patterning of CNS cells, myelination, and formation and refinement of neural circuits. Furthermore, we review studies suggesting that these cellular functions result in the modulation of behavior, which has important implications for a variety of neurological disorders.
Collapse
Affiliation(s)
- Jeffrey L Frost
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Dorothy P Schafer
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
359
|
Crotti A, Ransohoff RM. Microglial Physiology and Pathophysiology: Insights from Genome-wide Transcriptional Profiling. Immunity 2016; 44:505-515. [DOI: 10.1016/j.immuni.2016.02.013] [Citation(s) in RCA: 256] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/06/2016] [Accepted: 02/17/2016] [Indexed: 12/22/2022]
|
360
|
Hristovska I, Pascual O. Deciphering Resting Microglial Morphology and Process Motility from a Synaptic Prospect. Front Integr Neurosci 2016; 9:73. [PMID: 26834588 PMCID: PMC4717304 DOI: 10.3389/fnint.2015.00073] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/21/2015] [Indexed: 12/31/2022] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), were traditionally believed to be set into action only in case of injury or disease. Accordingly, microglia were assumed to be inactive or resting in the healthy brain. However, recent studies revealed that microglia carry out active tissue sampling in the intact brain by extending and retracting their ramified processes while periodically contacting synapses. Microglial morphology and motility as well as the frequency and duration of physical contacts with synaptic elements were found to be modulated by neuronal activity, sensory experience and neurotransmission; however findings have not been straightforward. Microglial cells are the most morphologically plastic element of the CNS. This unique feature confers them the possibility to locally sense activity, and to respond adequately by establishing synaptic contacts to regulate synaptic inputs by the secretion of signaling molecules. Indeed, microglial cells can hold new roles as critical players in maintaining brain homeostasis and regulating synaptic number, maturation and plasticity. For this reason, a better characterization of microglial cells and cues mediating neuron-to-microglia communication under physiological conditions may help advance our understanding of the microglial behavior and its regulation in the healthy brain. This review highlights recent findings on the instructive role of neuronal activity on microglial motility and microglia-synapse interactions, focusing on the main transmitters involved in this communication and including newly described communication at the tripartite synapse.
Collapse
Affiliation(s)
- Ines Hristovska
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research CenterLyon, France; Université Claude Bernard Lyon 1Lyon, France
| | - Olivier Pascual
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research CenterLyon, France; Université Claude Bernard Lyon 1Lyon, France
| |
Collapse
|
361
|
Kawanabe A, Okamura Y. Effects of unsaturated fatty acids on the kinetics of voltage-gated proton channels heterologously expressed in cultured cells. J Physiol 2016; 594:595-610. [PMID: 26563684 DOI: 10.1113/jp271274] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/28/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Arachidonic acid (AA) greatly enhances the activity of the voltage-gated proton (Hv) channel, although its mechanism of action and physiological function remain unclear. In the present study, we analysed the effects of AA on proton currents through Hv channels heterologously expressed in HEK293T cells. The dramatic increase in proton current amplitude elicited by AA was accompanied by accelerated activation kinetics and a leftward shift in the voltage-dependence of activation. Mutagenesis studies suggest the two aforementioned effects of AA reflect two distinct structural mechanisms. Application of phospholipase A2 , which liberates AA from phospholipids in the membrane, also enhances Hv channel activity, supporting the idea that AA modulates Hv channel activity within physiological contexts. Unsaturated fatty acids are key components of the biological membranes of all cells, and precursors of mediators for cell signalling. Arachidonic acid (AA) is an unsaturated fatty acid known to modulate the activities of various ion channels, including the voltage-gated proton (Hv) channel, which supports the rapid production of reactive oxygen species (ROS) in phagocytes through regulation of pH and membrane potential. However, the molecular mechanisms and physiological functions of the effects of AA on Hv channels remain unclear. In the present study, we report an electrophysiological analysis of the effects of AA on the mouse Hv channel (mHv1) heterologously expressed in HEK293T cells. Application of AA to excised inside-out patch membranes rapidly induced a robust increase in the amplitude of the proton current through mHv1. The current increase was accompanied by accelerated activation kinetics and a small leftward shift of the current-voltage relationship. In monomeric channels lacking the coiled-coil region of the channel protein, the shift in the current-voltage relationship was diminished but activation and deactivation remained accelerated. Studies with several AA derivatives showed that double bonds and hydrophilic head groups are essential for the effect of AA, although charge was not important. The application of phospholipase A2 (PLA2), which generates AA from cell membrane phospholipids, stimulated mHv1 activity to a similar extent as direct application of ∼ 20 μM AA, suggesting that endogenous AA may regulate Hv channel activity.
Collapse
Affiliation(s)
- Akira Kawanabe
- Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| | - Yasushi Okamura
- Integrative Physiology, Department of Physiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan.,Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
| |
Collapse
|
362
|
von Bernhardi R, Heredia F, Salgado N, Muñoz P. Microglia Function in the Normal Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:67-92. [PMID: 27714685 DOI: 10.1007/978-3-319-40764-7_4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The activation of microglia has been recognized for over a century by their morphological changes. Long slender microglia acquire a short sturdy ramified shape when activated. During the past 20 years, microglia have been accepted as an essential cellular component for understanding the pathogenic mechanism of many brain diseases, including neurodegenerative diseases. More recently, functional studies and imaging in mouse models indicate that microglia are active in the healthy central nervous system. It has become evident that microglia release several signal molecules that play key roles in the crosstalk among brain cells, i.e., astrocytes and oligodendrocytes with neurons, as well as with regulatory immune cells. Recent studies also reveal the heterogeneous nature of microglia diverse functions depending on development, previous exposure to stimulation events, brain region of residence, or pathological state. Subjects to approach by future research are still the unresolved questions regarding the conditions and mechanisms that render microglia protective, capable of preventing or reducing damage, or deleterious, capable of inducing or facilitating the progression of neuropathological diseases. This novel knowledge will certainly change our view on microglia as therapeutic target, shifting our goal from their general silencing to the generation of treatments able to change their activation pattern.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile.
| | - Florencia Heredia
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Nicole Salgado
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| | - Paola Muñoz
- Escuela de Medicina. Departamento de Neurología, Pontificia Universidad Católica de Chile, Marcoleta 391, Santiago, Chile
| |
Collapse
|
363
|
Li Y, Du X, Pei G, Du J, Zhao J. β-Arrestin1 regulates the morphology and dynamics of microglia in zebrafish in vivo. Eur J Neurosci 2015; 43:131-8. [PMID: 26354363 DOI: 10.1111/ejn.13065] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 01/24/2023]
Abstract
Microglia are the primary immune cells in the central nervous system. Microglia typically exist in a 'resting' state in the healthy brain, with ramified processes dynamically exploring the surrounding microenvironment. They become 'activated' under pathological conditions with marked changes in morphology. However, the regulation of their morphology dynamics remains poorly understood. Here, using in vivo time-lapse imaging and three-dimensional morphology analysis of microglia in intact zebrafish larvae, we found that β-arrestin1, a multifunctional protein involved in various signal transductions, cell-autonomously regulated the microglial morphology. Knockdown of β-arrestin1 increased the volume size and process number of microglia but reduced the deformation speed in the resting state. Meanwhile, β-arrestin1 down-regulation led to a high frequency of phagocytic behaviour of microglia. These defects were partially rescued by over-expressing human β-arrestin1 in microglia. Our study indicated that microglial dynamics in the resting state can be regulated cell-autonomously by β-arrestin1 signalling.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xufei Du
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Gang Pei
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiulin Du
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jian Zhao
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.,Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
364
|
Zeng WZ, Liu DS, Liu L, She L, Wu LJ, Xu TL. Activation of acid-sensing ion channels by localized proton transient reveals their role in proton signaling. Sci Rep 2015; 5:14125. [PMID: 26370138 PMCID: PMC4569896 DOI: 10.1038/srep14125] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 08/18/2015] [Indexed: 12/31/2022] Open
Abstract
Extracellular transients of pH alterations likely mediate signal transduction in the nervous system. Neuronal acid-sensing ion channels (ASICs) act as sensors for extracellular protons, but the mechanism underlying ASIC activation remains largely unknown. Here, we show that, following activation of a light-activated proton pump, Archaerhodopsin-3 (Arch), proton transients induced ASIC currents in both neurons and HEK293T cells co-expressing ASIC1a channels. Using chimera proteins that bridge Arch and ASIC1a by a glycine/serine linker, we found that successful coupling occurred within 15 nm distance. Furthermore, two-cell sniffer patch recording revealed that regulated release of protons through either Arch or voltage-gated proton channel Hv1 activated neighbouring cells expressing ASIC1a channels. Finally, computational modelling predicted the peak proton concentration at the intercellular interface to be at pH 6.7, which is acidic enough to activate ASICs in vivo. Our results highlight the pathophysiological role of proton signalling in the nervous system.
Collapse
Affiliation(s)
- Wei-Zheng Zeng
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Di-Shi Liu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lu Liu
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang She
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Long-Jun Wu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Tian-Le Xu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
365
|
Choi I, Kim B, Byun JW, Baik SH, Huh YH, Kim JH, Mook-Jung I, Song WK, Shin JH, Seo H, Suh YH, Jou I, Park SM, Kang HC, Joe EH. LRRK2 G2019S mutation attenuates microglial motility by inhibiting focal adhesion kinase. Nat Commun 2015; 6:8255. [PMID: 26365310 PMCID: PMC4647842 DOI: 10.1038/ncomms9255] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 08/03/2015] [Indexed: 01/20/2023] Open
Abstract
In response to brain injury, microglia rapidly extend processes that isolate lesion sites and protect the brain from further injury. Here we report that microglia carrying a pathogenic mutation in the Parkinson's disease (PD)-associated gene, G2019S-LRRK2 (GS-Tg microglia), show retarded ADP-induced motility and delayed isolation of injury, compared with non-Tg microglia. Conversely, LRRK2 knockdown microglia are highly motile compared with control cells. In our functional assays, LRRK2 binds to focal adhesion kinase (FAK) and phosphorylates its Thr–X–Arg/Lys (TXR/K) motif(s), eventually attenuating FAK activity marked by decreased pY397 phosphorylation (pY397). GS-LRRK2 decreases the levels of pY397 in the brain, microglia and HEK cells. In addition, treatment with an inhibitor of LRRK2 kinase restores pY397 levels, decreased pTXR levels and rescued motility of GS-Tg microglia. These results collectively suggest that G2019S mutation of LRRK2 may contribute to the development of PD by inhibiting microglial response to brain injury. In response to brain injury, microglia extend processes to isolate the lesion. Here Choi et al. show that microglia expressing a pathogenic mutation in the Parkinson's disease-associated LRRK2 gene show reduced motility and delayed lesion isolation in vitro and in vivo due to attenuated focal adhesion kinase activity.
Collapse
Affiliation(s)
- Insup Choi
- Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Department of Pharmacology, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea
| | - Beomsue Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea
| | - Ji-Won Byun
- Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Department of Pharmacology, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea
| | - Sung Hoon Baik
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Korea
| | - Yun Hyun Huh
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | - Jong-Hyeon Kim
- Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Department of Pharmacology, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Korea
| | - Woo Keun Song
- Bio Imaging and Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | - Joo-Ho Shin
- Division of Pharmacology, Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 440-746, Korea
| | - Hyemyung Seo
- Department of Molecular and Life Sciences, Hanyang University, Ansan 426-791, Korea
| | - Young Ho Suh
- Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Department of Pharmacology, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea
| | - Ilo Jou
- Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Department of Pharmacology, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea
| | - Sang Myun Park
- Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Department of Pharmacology, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea
| | - Ho Chul Kang
- Department of Physiology, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea
| | - Eun-Hye Joe
- Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Department of Pharmacology, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea.,Brain Disease Research Center, Ajou University School of Medicine, Suwon, Gyeonggi-do 443-380, Korea
| |
Collapse
|
366
|
Morsch M, Radford R, Lee A, Don EK, Badrock AP, Hall TE, Cole NJ, Chung R. In vivo characterization of microglial engulfment of dying neurons in the zebrafish spinal cord. Front Cell Neurosci 2015; 9:321. [PMID: 26379496 PMCID: PMC4553390 DOI: 10.3389/fncel.2015.00321] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/03/2015] [Indexed: 11/13/2022] Open
Abstract
Microglia are specialized phagocytes in the vertebrate central nervous system (CNS). As the resident immune cells of the CNS they play an important role in the removal of dying neurons during both development and in several neuronal pathologies. Microglia have been shown to prevent the diffusion of damaging degradation products of dying neurons by engulfment and ingestion. Here we describe a live imaging approach that uses UV laser ablation to selectively stress and kill spinal neurons and visualize the clearance of neuronal remnants by microglia in the zebrafish spinal cord. In vivo imaging confirmed the motile nature of microglia within the uninjured spinal cord. However, selective neuronal ablation triggered rapid activation of microglia, leading to phagocytic uptake of neuronal debris by microglia within 20-30 min. This process of microglial engulfment is highly dynamic, involving the extension of processes toward the lesion site and consequently the ingestion of the dying neuron. 3D rendering analysis of time-lapse recordings revealed the formation of phagosome-like structures in the activated microglia located at the site of neuronal ablation. This real-time representation of microglial phagocytosis in the living zebrafish spinal cord provides novel opportunities to study the mechanisms of microglia-mediated neuronal clearance.
Collapse
Affiliation(s)
- Marco Morsch
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Rowan Radford
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Albert Lee
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Emily K Don
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Andrew P Badrock
- Faculty of Life Sciences, The University of Manchester Manchester, UK
| | - Thomas E Hall
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland Brisbane, QLD, Australia
| | - Nicholas J Cole
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Roger Chung
- Motor Neuron Disease Research Group, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| |
Collapse
|
367
|
Wei R, Wang J, Xu Y, Yin B, He F, Du Y, Peng G, Luo B. Probenecid protects against cerebral ischemia/reperfusion injury by inhibiting lysosomal and inflammatory damage in rats. Neuroscience 2015; 301:168-77. [PMID: 26047730 DOI: 10.1016/j.neuroscience.2015.05.070] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/14/2015] [Accepted: 05/28/2015] [Indexed: 10/23/2022]
Abstract
Probenecid has been used for decades to treat gout, and recent studies have revealed it is also a specific inhibitor of the pannexin-1 channel. It has been reported that the pannexin-1 channel is involved in ischemic injury. Here, we investigated the neuroprotective effect and the possible mechanisms of action of probenecid in global cerebral ischemia/reperfusion (I/R) injury in rats. Twenty minutes of transient global cerebral I/R injury was induced using the four-vessel occlusion (4-VO) method in male Sprague-Dawley rats. Different doses of probenecid were administered intravenously, intraperitoneally, or by gavage before or after reperfusion. Probenecid via all three routes protected against CA1 neuronal death when given before reperfusion. This protective effect continued when probenecid was given at 2h after reperfusion, but not at 6h. Interestingly, the protective effect regained if probenecid was given continuously for 7days after reperfusion. The release of cathepsin B and overexpression of calpain-1 after reperfusion were inhibited, while the upregulation of Hsp70 was strengthened by probenecid pre-treatment. Furthermore, the activation and proliferation of microglia and astrocytes after I/R injury were suppressed by continuous given for 7days, but only partly by a single dose at 6h of reperfusion. Thus, our data indicate that probenecid protects against transient global cerebral I/R injury probably by inhibiting calpain-cathepsin pathway and the inflammatory reaction.
Collapse
Affiliation(s)
- R Wei
- The Brain Medical Center and the Collaborative Innovation Center for Brain Science, Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - J Wang
- Department of Neurology, The First Affiliated Hospital, Anhui Medical University, Hefei 230022, China
| | - Y Xu
- The Brain Medical Center and the Collaborative Innovation Center for Brain Science, Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - B Yin
- Department of Neurology, Renmin Hospital, Wuhan University, Wuhan 430000, China
| | - F He
- The Brain Medical Center and the Collaborative Innovation Center for Brain Science, Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Y Du
- The Brain Medical Center and the Collaborative Innovation Center for Brain Science, Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - G Peng
- The Brain Medical Center and the Collaborative Innovation Center for Brain Science, Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - B Luo
- The Brain Medical Center and the Collaborative Innovation Center for Brain Science, Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
368
|
TNF and its receptors in the CNS: The essential, the desirable and the deleterious effects. Neuroscience 2015; 302:2-22. [DOI: 10.1016/j.neuroscience.2015.06.038] [Citation(s) in RCA: 358] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/17/2015] [Accepted: 06/19/2015] [Indexed: 12/15/2022]
|
369
|
Morara S, Colangelo AM, Provini L. Microglia-Induced Maladaptive Plasticity Can Be Modulated by Neuropeptides In Vivo. Neural Plast 2015; 2015:135342. [PMID: 26273481 PMCID: PMC4529944 DOI: 10.1155/2015/135342] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/25/2015] [Indexed: 02/06/2023] Open
Abstract
Microglia-induced maladaptive plasticity is being recognized as a major cause of deleterious self-sustaining pathological processes that occur in neurodegenerative and neuroinflammatory diseases. Microglia, the primary homeostatic guardian of the central nervous system, exert critical functions both during development, in neural circuit reshaping, and during adult life, in the brain physiological and pathological surveillance. This delicate critical role can be disrupted by neural, but also peripheral, noxious stimuli that can prime microglia to become overreactive to a second noxious stimulus or worsen underlying pathological processes. Among regulators of microglia, neuropeptides can play a major role. Their receptors are widely expressed in microglial cells and neuropeptide challenge can potently influence microglial activity in vitro. More relevantly, this regulator activity has been assessed also in vivo, in experimental models of brain diseases. Neuropeptide action in the central nervous system has been associated with beneficial effects in neurodegenerative and neuroinflammatory pathological experimental models. This review describes some of the mechanisms of the microglia maladaptive plasticity in vivo and how neuropeptide activity can represent a useful therapeutical target in a variety of human brain pathologies.
Collapse
Affiliation(s)
- Stefano Morara
- Neuroscience Institute (CNR), Via Vanvitelli 32, 20129 Milano, Italy
- Department of BIOMETRA, University of Milano, Via Vanvitelli 32, 20129 Milano, Italy
| | - Anna Maria Colangelo
- Laboratory of Neuroscience “R. Levi-Montalcini”, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy
- SYSBIO Centre of Systems Biology, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milano, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, 20126 Milano, Italy
| | - Luciano Provini
- Department of BIOMETRA, University of Milano, Via Vanvitelli 32, 20129 Milano, Italy
| |
Collapse
|
370
|
Squarzoni P, Thion MS, Garel S. Neuronal and microglial regulators of cortical wiring: usual and novel guideposts. Front Neurosci 2015; 9:248. [PMID: 26236185 PMCID: PMC4505395 DOI: 10.3389/fnins.2015.00248] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022] Open
Abstract
Neocortex functioning relies on the formation of complex networks that begin to be assembled during embryogenesis by highly stereotyped processes of cell migration and axonal navigation. The guidance of cells and axons is driven by extracellular cues, released along by final targets or intermediate targets located along specific pathways. In particular, guidepost cells, originally described in the grasshopper, are considered discrete, specialized cell populations located at crucial decision points along axonal trajectories that regulate tract formation. These cells are usually early-born, transient and act at short-range or via cell-cell contact. The vast majority of guidepost cells initially identified were glial cells, which play a role in the formation of important axonal tracts in the forebrain, such as the corpus callosum, anterior, and post-optic commissures as well as optic chiasm. In the last decades, tangential migrating neurons have also been found to participate in the guidance of principal axonal tracts in the forebrain. This is the case for several examples such as guideposts for the lateral olfactory tract (LOT), corridor cells, which open an internal path for thalamo-cortical axons and Cajal-Retzius cells that have been involved in the formation of the entorhino-hippocampal connections. More recently, microglia, the resident macrophages of the brain, were specifically observed at the crossroads of important neuronal migratory routes and axonal tract pathways during forebrain development. We furthermore found that microglia participate to the shaping of prenatal forebrain circuits, thereby opening novel perspectives on forebrain development and wiring. Here we will review the last findings on already known guidepost cell populations and will discuss the role of microglia as a potentially new class of atypical guidepost cells.
Collapse
Affiliation(s)
- Paola Squarzoni
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Morgane S Thion
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Sonia Garel
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| |
Collapse
|
371
|
Kolodziejczak M, Béchade C, Gervasi N, Irinopoulou T, Banas SM, Cordier C, Rebsam A, Roumier A, Maroteaux L. Serotonin Modulates Developmental Microglia via 5-HT2B Receptors: Potential Implication during Synaptic Refinement of Retinogeniculate Projections. ACS Chem Neurosci 2015; 6:1219-30. [PMID: 25857335 DOI: 10.1021/cn5003489] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Maturation of functional neuronal circuits during central nervous system development relies on sophisticated mechanisms. First, axonal and dendritic growth should reach appropriate targets for correct synapse elaboration. Second, pruning and neuronal death are required to eliminate redundant or inappropriate neuronal connections. Serotonin, in addition to its role as a neurotransmitter, actively participates in postnatal establishment and refinement of brain wiring in mammals. Brain resident macrophages, that is, microglia, also play an important role in developmentally regulated neuronal death as well as in synaptic maturation and elimination. Here, we tested the hypothesis of cross-regulation between microglia and serotonin during postnatal brain development in a mouse model of synaptic refinement. We found expression of the serotonin 5-HT2B receptor on postnatal microglia, suggesting that serotonin could participate in temporal and spatial synchronization of microglial functions. Using two-photon microscopy, acute brain slices, and local delivery of serotonin, we observed that microglial processes moved rapidly toward the source of serotonin in Htr2B(+/+) mice, but not in Htr2B(-/-) mice lacking the 5-HT2B receptor. We then investigated whether some developmental steps known to be controlled by serotonin could potentially result from microglia sensitivity to serotonin. Using an in vivo model of synaptic refinement during early brain development, we investigated the maturation of the retinal projections to the thalamus and observed that Htr2B(-/-) mice present anatomical alterations of the ipsilateral projecting area of retinal axons into the thalamus. In addition, activation markers were upregulated in microglia from Htr2B(-/-) compared to control neonates, in the absence of apparent morphological modifications. These results support the hypothesis that serotonin interacts with microglial cells and these interactions participate in brain maturation.
Collapse
Affiliation(s)
- Marta Kolodziejczak
- INSERM UMR-S 839, F75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F75005, Paris, France
- Institut du Fer à Moulin, 17 rue du Fer à Moulin, F75005, Paris, France
| | - Catherine Béchade
- INSERM UMR-S 839, F75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F75005, Paris, France
- Institut du Fer à Moulin, 17 rue du Fer à Moulin, F75005, Paris, France
| | - Nicolas Gervasi
- INSERM UMR-S 839, F75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F75005, Paris, France
- Institut du Fer à Moulin, 17 rue du Fer à Moulin, F75005, Paris, France
| | - Theano Irinopoulou
- INSERM UMR-S 839, F75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F75005, Paris, France
- Institut du Fer à Moulin, 17 rue du Fer à Moulin, F75005, Paris, France
| | - Sophie M. Banas
- INSERM UMR-S 839, F75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F75005, Paris, France
- Institut du Fer à Moulin, 17 rue du Fer à Moulin, F75005, Paris, France
| | - Corinne Cordier
- Cytometry Facility, INSERM US24, F75005, Paris, France
- CNRS UMS 3633, Paris Descartes University, F75005, Paris, France
| | - Alexandra Rebsam
- INSERM UMR-S 839, F75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F75005, Paris, France
- Institut du Fer à Moulin, 17 rue du Fer à Moulin, F75005, Paris, France
| | - Anne Roumier
- INSERM UMR-S 839, F75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F75005, Paris, France
- Institut du Fer à Moulin, 17 rue du Fer à Moulin, F75005, Paris, France
| | - Luc Maroteaux
- INSERM UMR-S 839, F75005, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, F75005, Paris, France
- Institut du Fer à Moulin, 17 rue du Fer à Moulin, F75005, Paris, France
| |
Collapse
|
372
|
Kälin S, Heppner FL, Bechmann I, Prinz M, Tschöp MH, Yi CX. Hypothalamic innate immune reaction in obesity. Nat Rev Endocrinol 2015; 11:339-51. [PMID: 25824676 DOI: 10.1038/nrendo.2015.48] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Findings from rodent and human studies show that the presence of inflammatory factors is positively correlated with obesity and the metabolic syndrome. Obesity-associated inflammatory responses take place not only in the periphery but also in the brain. The hypothalamus contains a range of resident glial cells including microglia, macrophages and astrocytes, which are embedded in highly heterogenic groups of neurons that control metabolic homeostasis. This complex neural-glia network can receive information directly from blood-borne factors, positioning it as a metabolic sensor. Following hypercaloric challenge, mediobasal hypothalamic microglia and astrocytes enter a reactive state, which persists during diet-induced obesity. In established mouse models of diet-induced obesity, the hypothalamic vasculature displays angiogenic alterations. Moreover, proopiomelanocortin neurons, which regulate food intake and energy expenditure, are impaired in the arcuate nucleus, where there is an increase in local inflammatory signals. The sum total of these events is a hypothalamic innate immune reactivity, which includes temporal and spatial changes to each cell population. Although the exact role of each participant of the neural-glial-vascular network is still under exploration, therapeutic targets for treating obesity should probably be linked to individual cell types and their specific signalling pathways to address each dysfunction with cell-selective compounds.
Collapse
Affiliation(s)
- Stefanie Kälin
- Institute for Diabetes and Obesity, Helmholtz Centre for Health and Environment &Technische Universität München, 85748, Munich, Germany
| | - Frank L Heppner
- Department of Neuropathology, Charité, Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Liebigstr. 13, 04103 Leipzig, Germany
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, Breisacher Str. 64, D-79106 Freiburg, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Centre for Health and Environment &Technische Universität München, 85748, Munich, Germany
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, Netherlands
| |
Collapse
|
373
|
TLR1 expression in mouse brain was increased in a KA-induced seizure model. Inflamm Res 2015; 64:487-95. [PMID: 26021825 DOI: 10.1007/s00011-015-0828-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 04/16/2015] [Accepted: 04/23/2015] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Toll-like receptors (TLRs) that mediate inflammatory responses play an important role in epilepsy; however, whether TLR1 is also involved in epileptogenesis remains unclear. Thus, in this study, we investigated the extent and pattern of TLR1 expression in epileptic tissues. METHODS One-hundred and thirty-two mice were intra-cerebroventricularly injected with PBS or kainic acid (KA) and were examined at 1, 3, 8 and 24 h. The expression pattern and distribution of TLR1 were examined by reverse-transcriptase polymerase chain reaction (RT-PCR), western blot analysis and immunohistochemistry staining. RESULTS The mRNA and protein levels of TLR1 were significantly upregulated in the hippocampus and temporal cortex of epileptic mice compared with those of controls. TLR1 expression was increased as early as 1 h following KA treatment and peaked at 8 and 24 h. Immunohistochemistry staining demonstrated that TLR1 was distributed in the CA1-3, dentate gyrus and hilus regions of the hippocampus and different cortical regions. Immunofluorescent staining further revealed that TLR1 was primarily expressed in the neurons, microglia, and astrocytes of epileptogenic tissue. SIGNIFICANCE These results demonstrate that cortical and hippocampal sub-regional expression of TLR1 is altered during epileptogenesis in a time- and location-specific manner, suggesting a close association with the process of epilepsy.
Collapse
|
374
|
Abstract
Microglia are macrophages that colonize the brain during development to establish a resident population of professional phagocytes that protect against invading pathogens and contribute to brain development and homeostasis. As such, these cells sit at the interface between immunology and neurobiology. In addition to their key roles in brain physiology, microglia offer a great opportunity to address central questions in biology relating to how migrating cells find their positions in the embryo, adopt a behavior that is appropriate for that position, and interact with their local environment. We aim, in this review, to survey key recent advances in microglial research.
Collapse
Affiliation(s)
- Alessandra Maria Casano
- European Molecular Biology Laboratory, Developmental Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Francesca Peri
- European Molecular Biology Laboratory, Developmental Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany.
| |
Collapse
|
375
|
Modulation of microglial process convergence toward neuronal dendrites by extracellular calcium. J Neurosci 2015; 35:2417-22. [PMID: 25673836 DOI: 10.1523/jneurosci.3279-14.2015] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Extracellular calcium concentrations in the brain fluctuate during neuronal activities and may affect the behavior of brain cells. Microglia are highly dynamic immune cells of the brain. However, the effects of extracellular calcium concentrations on microglial dynamics have not been investigated. Here, we addressed this question in mouse brain slices and in vivo using two-photon microscopy. We serendipitously found that extracellular calcium reduction induced microglial processes to converge at distinct sites, a phenomenon we termed microglial process convergence (MPCs). Our studies revealed that MPCs target neuronal dendrites independent of neuronal action potential firing and is mediated by ATP release and microglial P2Y12 receptors. These results indicate that microglia monitor and interact with neurons during conditions of cerebral calcium reduction in the normal and diseased brain.
Collapse
|
376
|
Abstract
Microglia are the resident immune cells in the CNS and play diverse roles in the maintenance of CNS homeostasis. Recent studies have shown that microglia continually survey the CNS microenvironment and scavenge cell debris and aberrant proteins by phagocytosis and pinocytosis, and that reactive microglia are capable to present antigens to T cells and initiate immune responses. However, how microglia process the endocytosed contents and evoke an immune response remain unclear. Here we report that a size-dependent selective transport of small soluble contents from the pinosomal lumen into lysosomes is critical for the antigen processing in microglia. Using fluorescent probes and water-soluble magnetic nanobeads of defined sizes, we showed in cultured rodent microglia, and in a cell-free reconstructed system that pinocytosed proteins become degraded immediately following pinocytosis and the resulting peptides are selectively delivered to major histocompatibility complex class II (MHC-II) containing lysosomes, whereas undegraded proteins are retained in the pinosomal lumen. This early size-based sorting of pinosomal contents relied on the formation of transient tunnel between pinosomes and lysosomes in a Rab7- and dynamin II-dependent manner, which allowed the small contents to pass through but restricted large ones. Inhibition of the size-based sorting markedly reduced proliferation and cytokine release of cocultured CD4(+) T cells, indicating that the size-based sorting is required for efficient antigen presentation by microglial cells. Together, these findings reveal a novel early sorting mechanism for pinosomal luminal contents in microglial cells, which may explain how microglia efficiently process protein antigens and evoke an immune response.
Collapse
|
377
|
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: 68] [Impact Index Per Article: 6.8] [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
|
378
|
George J, Gonçalves FQ, Cristóvão G, Rodrigues L, Meyer Fernandes JR, Gonçalves T, Cunha RA, Gomes CA. Different danger signals differently impact on microglial proliferation through alterations of ATP release and extracellular metabolism. Glia 2015; 63:1636-45. [PMID: 25847308 DOI: 10.1002/glia.22833] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 03/17/2015] [Indexed: 12/20/2022]
Abstract
Microglia rely on their ability to proliferate in the brain parenchyma to sustain brain innate immunity and participate in the reaction to brain damage. We now studied the influence of different danger signals activating microglia, both internal (typified by glutamate, associated with brain damage) and external (using a bacterial lipopolysaccharide, LPS), on the proliferation of microglia cells. We found that LPS (100 ng/mL) increased, whereas glutamate (0.5 mM) decreased proliferation. Notably, LPS decreased whereas glutamate increased the extracellular levels of ATP. In contrast, LPS increased whereas glutamate decreased the extracellular catabolism of ATP into adenosine through ecto-nucleotidases and ecto-5'-nucleotidase. Finally, apyrase (degrades extracellular ATP) abrogated glutamate-induced inhibition of microglia proliferation; conversely, inhibitors of ecto-nucleotidases (ARL67156 or α,β-methylene ADP) and adenosine deaminase (degrades extracellular adenosine) abrogated the LPS-induced increase of microglia proliferation, which was blocked by a selective A2A receptor antagonist, SCH58261 (50 nM). Overall, these results highlight the importance of the extracellular purinergic metabolism to format microglia proliferation and influence the spatio-temporal profile of neuroinflammation in different conditions of brain damage.
Collapse
Affiliation(s)
- Jimmy George
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | | | - Gonçalo Cristóvão
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Lisa Rodrigues
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal.,FMUC-Faculty of Medicine, University of Coimbra, Portugal
| | | | - Teresa Gonçalves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal.,FMUC-Faculty of Medicine, University of Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal.,FMUC-Faculty of Medicine, University of Coimbra, Portugal
| | - Catarina A Gomes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal.,FMUC-Faculty of Medicine, University of Coimbra, Portugal
| |
Collapse
|
379
|
Madeira MH, Boia R, Santos PF, Ambrósio AF, Santiago AR. Contribution of microglia-mediated neuroinflammation to retinal degenerative diseases. Mediators Inflamm 2015; 2015:673090. [PMID: 25873768 PMCID: PMC4385698 DOI: 10.1155/2015/673090] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/16/2014] [Indexed: 12/27/2022] Open
Abstract
Retinal degenerative diseases are major causes of vision loss and blindness worldwide and are characterized by chronic and progressive neuronal loss. One common feature of retinal degenerative diseases and brain neurodegenerative diseases is chronic neuroinflammation. There is growing evidence that retinal microglia, as in the brain, become activated in the course of retinal degenerative diseases, having a pivotal role in the initiation and propagation of the neurodegenerative process. A better understanding of the events elicited and mediated by retinal microglia will contribute to the clarification of disease etiology and might open new avenues for potential therapeutic interventions. This review aims at giving an overview of the roles of microglia-mediated neuroinflammation in major retinal degenerative diseases like glaucoma, age-related macular degeneration, and diabetic retinopathy.
Collapse
Affiliation(s)
- Maria H. Madeira
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
| | - Raquel Boia
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
| | - Paulo F. Santos
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - António F. Ambrósio
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- AIBILI, Coimbra, Portugal
| | - Ana R. Santiago
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- AIBILI, Coimbra, Portugal
| |
Collapse
|
380
|
Engl E, Attwell D. Non-signalling energy use in the brain. J Physiol 2015; 593:3417-29. [PMID: 25639777 PMCID: PMC4560575 DOI: 10.1113/jphysiol.2014.282517] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 01/27/2015] [Indexed: 01/19/2023] Open
Abstract
Energy use limits the information processing power of the brain. However, apart from the ATP used to power electrical signalling, a significant fraction of the brain's energy consumption is not directly related to information processing. The brain spends just under half of its energy on non-signalling processes, but it remains poorly understood which tasks are so energetically costly for the brain. We review existing experimental data on subcellular processes that may contribute to this non-signalling energy use, and provide modelling estimates, to try to assess the magnitude of their ATP consumption and consider how their changes in pathology may compromise neuronal function. As a main result, surprisingly little consensus exists on the energetic cost of actin treadmilling, with estimates ranging from < 1% of the brain's global energy budget up to one-half of neuronal energy use. Microtubule treadmilling and protein synthesis have been estimated to account for very small fractions of the brain's energy budget, whereas there is stronger evidence that lipid synthesis and mitochondrial proton leak are energetically expensive. Substantial further research is necessary to close these gaps in knowledge about the brain's energy-expensive non-signalling tasks.
Collapse
Affiliation(s)
- Elisabeth Engl
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, WC1E 6BT, UK
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, WC1E 6BT, UK
| |
Collapse
|
381
|
Chen Z, Trapp BD. Microglia and neuroprotection. J Neurochem 2015; 136 Suppl 1:10-7. [PMID: 25693054 DOI: 10.1111/jnc.13062] [Citation(s) in RCA: 298] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/29/2015] [Accepted: 02/02/2015] [Indexed: 12/11/2022]
Abstract
Microglia were first identified over a century ago, but our knowledge about their ontogeny and functions has significantly expanded only recently. Microglia colonize the central nervous system (CNS) in utero and play essential roles in brain development. Once neural development is completed, microglia function as the resident innate immune cells of the CNS by surveying their microenvironment and becoming activated when the CNS is challenged by infection, injury, or disease. Despite the traditional view of microglia as being destructive in neurological diseases, recent studies have shown that microglia maintain CNS homeostasis and protect the CNS under various pathological conditions. Microglia can be prophylactically activated by modeling infection with systemic lipopolysaccharide injections and these activated microglia can protect the brain from traumatic injury through modulation of neuronal synapses. Microglia can also protect the CNS by promoting neurogenesis, clearing debris, and suppressing inflammation in diseases such as stroke, autism, and Alzheimer's. Microglia are the resident innate immune cells of the CNS. Despite the traditional view of microglia as being destructive in neurological diseases, recent studies have shown that they maintain tissue homeostasis and protect the CNS under various pathological conditions. They achieve so by clearing debris, promoting neurogenesis, suppressing inflammation and stripping inhibitory synapses. This review summarizes recent advances of our understanding on the multi-dimensional neuroprotective roles of microglia.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
382
|
Almolda B, de Labra C, Barrera I, Gruart A, Delgado-Garcia JM, Villacampa N, Vilella A, Hofer MJ, Hidalgo J, Campbell IL, González B, Castellano B. Alterations in microglial phenotype and hippocampal neuronal function in transgenic mice with astrocyte-targeted production of interleukin-10. Brain Behav Immun 2015; 45:80-97. [PMID: 25449577 DOI: 10.1016/j.bbi.2014.10.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/24/2014] [Accepted: 10/25/2014] [Indexed: 12/31/2022] Open
Abstract
Interleukin-10 (IL-10) is a cytokine classically linked with anti-inflammatory and protective functions in the central nervous system (CNS) in different neurodegenerative and neuroinflammatory conditions. In order to study the specific role of local CNS produced IL-10, we have created a new transgenic mouse line with astrocyte-targeted production of IL-10 (GFAP-IL10Tg). In the present study, the effects of local CNS IL-10 production on microglia, astrocytes and neuronal connectivity under basal conditions were investigated using immunohistochemistry, molecular biology techniques, electrophysiology and behavioural studies. Our results showed that, in GFAP-IL10Tg animals, microglia displayed an increase in density and a specific activated phenotype characterised by morphological changes in specific areas of the brain including the hippocampus, cortex and cerebellum that correlated with the level of transgene expressed IL-10 mRNA. Distinctively, in the hippocampus, microglial cells adopted an elongated morphology following the same direction as the dendrites of pyramidal neurons. Moreover, this IL-10-induced microglial phenotype showed increased expression of certain molecules including Iba1, CD11b, CD16/32 and F4/80 markers, "de novo" expression of CD150 and no detectable levels of either CD206 or MHCII. To evaluate whether this specific activated microglial phenotype was associated with changes in neuronal activity, the electrophysiological properties of pyramidal neurons of the hippocampus (CA3-CA1) were analysed in vivo. We found a lower excitability of the CA3-CA1 synapses and absence of long-term potentiation (LTP) in GFAP-IL10Tg mice. This study is the first description of a transgenic mouse with astrocyte-targeted production of the cytokine IL-10. The findings indicate that IL-10 induces a specific activated microglial phenotype concomitant with changes in hippocampal LTP responses. This transgenic animal will be a very useful tool to study IL-10 functions in the CNS, not only under basal conditions, but also after different experimental lesions or induced diseases.
Collapse
Affiliation(s)
- Beatriz Almolda
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain.
| | - Carmen de Labra
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Iliana Barrera
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Agnès Gruart
- Division of Neurosciences, Pablo de Olavide University, Seville 41013, Spain
| | | | - Nàdia Villacampa
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Antonietta Vilella
- Department of Biomedical, Metabolic and Neural Sciences, Università degli Studi di Modena e Reggio Emilia, 41125, Italy
| | - Markus J Hofer
- School of Molecular Bioscience, The University of Sydney, Sydney, NSW 2006, Australia
| | - Juan Hidalgo
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Iain L Campbell
- School of Molecular Bioscience, The University of Sydney, Sydney, NSW 2006, Australia
| | - Berta González
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Bernardo Castellano
- Department of Cell Biology, Physiology and Immunology, Institute of Neuroscience, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| |
Collapse
|
383
|
Moritz C, Berardi F, Abate C, Peri F. Live imaging reveals a new role for the sigma-1 (σ1) receptor in allowing microglia to leave brain injuries. Neurosci Lett 2015; 591:13-18. [PMID: 25666889 DOI: 10.1016/j.neulet.2015.02.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/20/2015] [Accepted: 02/03/2015] [Indexed: 12/21/2022]
Abstract
Microglial cells are responsible for clearing and maintaining the central nervous system (CNS) microenvironment. Upon brain damage, they move toward injuries to clear the area by engulfing dying neurons. However, in the context of many neurological disorders chronic microglial responses are responsible for neurodegeneration. Therefore, it is important to understand how these cells can be "switched-off" and regain their ramified state. Current research suggests that microglial inflammatory responses can be inhibited by sigma (σ) receptor activation. Here, we take advantage of the optical transparency of the zebrafish embryo to study the role of σ1 receptor in microglia in an intact living brain. By combining chemical approaches with real time imaging we found that treatment with PB190, a σ1 agonist, blocks microglial migration toward injuries leaving cellular baseline motility and the engulfment of apoptotic neurons unaffected. Most importantly, by taking a reverse genetic approach, we discovered that the role of σ1in vivo is to "switch-off" microglia after they responded to an injury allowing for these cells to leave the site of damage. This indicates that pharmacological manipulation of σ1 receptor modulates microglial responses providing new approaches to reduce the devastating impact that microglia have in neurodegenerative diseases.
Collapse
Affiliation(s)
- Christian Moritz
- European Molecular Biology Laboratory, Developmental Biology Unit, Meyerhofstr. 1, 69117 Heidelberg, Germany
| | - Francesco Berardi
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, I-70125 Bari, Italy
| | - Carmen Abate
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, I-70125 Bari, Italy.
| | - Francesca Peri
- European Molecular Biology Laboratory, Developmental Biology Unit, Meyerhofstr. 1, 69117 Heidelberg, Germany.
| |
Collapse
|
384
|
Baalman K, Marin MA, Ho TSY, Godoy M, Cherian L, Robertson C, Rasband MN. Axon initial segment-associated microglia. J Neurosci 2015; 35:2283-92. [PMID: 25653382 PMCID: PMC4315845 DOI: 10.1523/jneurosci.3751-14.2015] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/02/2014] [Accepted: 12/22/2014] [Indexed: 01/31/2023] Open
Abstract
Microglia are the brain's resident immune cells and function as the main defense against pathogens or injury. However, in the absence of disease, microglia have other functions in the normal brain. For example, previous studies showed that microglia contribute to circuit refinement and synaptic plasticity in the developing and adult brain, respectively. Thus, microglia actively participate in regulating neuronal excitability and function. Here, we report that in the cortex, but not other brain regions, a subset of microglia extend a single process that specifically associates and overlaps with the axon initial segment (AIS), the site where action potentials are generated. Similar associations were not observed with dendrites or distal axons. Microglia-AIS interactions appear early in development, persist throughout adulthood, and are conserved across species including mice, rats, and primates. However, these interactions are lost after microglial activation following brain injury, suggesting that such interactions may be part of healthy brain function. Loss of microglial CX3CR1 receptors, or the specialized extracellular matrix surrounding the AIS, did not disrupt the interaction. However, loss of AIS proteins by the neuron-specific deletion of the master AIS scaffold AnkyrinG disrupted microglia-AIS interactions. These results reveal a unique population of microglia that specifically interact with the AIS in the adult cortex.
Collapse
Affiliation(s)
| | | | | | | | - Leela Cherian
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030
| | - Claudia Robertson
- Department of Neuroscience, Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030
| | | |
Collapse
|
385
|
Delpech JC, Madore C, Nadjar A, Joffre C, Wohleb ES, Layé S. Microglia in neuronal plasticity: Influence of stress. Neuropharmacology 2015; 96:19-28. [PMID: 25582288 DOI: 10.1016/j.neuropharm.2014.12.034] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 12/24/2014] [Accepted: 12/29/2014] [Indexed: 01/17/2023]
Abstract
The central nervous system (CNS) has previously been regarded as an immune-privileged site with the absence of immune cell responses but this dogma was not entirely true. Microglia are the brain innate immune cells and recent findings indicate that they participate both in CNS disease and infection as well as facilitate normal CNS function. Microglia are highly plastic and play integral roles in sculpting the structure of the CNS, refining neuronal circuitry and connectivity, and contribute actively to neuronal plasticity in the healthy brain. Interestingly, psychological stress can perturb the function of microglia in association with an impaired neuronal plasticity and the development of emotional behavior alterations. As a result it seemed important to describe in this review some findings indicating that the stress-induced microglia dysfunction may underlie neuroplasticity deficits associated to many mood disorders. This article is part of a Special Issue entitled 'Neuroimmunology and Synaptic Function'.
Collapse
Affiliation(s)
- Jean-Christophe Delpech
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France
| | - Charlotte Madore
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France
| | - Agnes Nadjar
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France
| | - Corinne Joffre
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France
| | - Eric S Wohleb
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Sophie Layé
- Nutrition et Neurobiologie Intégrée, INRA 1286, 33077 Bordeaux Cedex, France; Nutrition et Neurobiologie Intégrée, University of Bordeaux, Bordeaux 33077, France.
| |
Collapse
|
386
|
|
387
|
Blackiston DJ, Anderson GM, Rahman N, Bieck C, Levin M. A novel method for inducing nerve growth via modulation of host resting potential: gap junction-mediated and serotonergic signaling mechanisms. Neurotherapeutics 2015; 12:170-84. [PMID: 25449797 PMCID: PMC4322068 DOI: 10.1007/s13311-014-0317-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A major goal of regenerative medicine is to restore the function of damaged or missing organs through the implantation of bioengineered or donor-derived components. It is necessary to understand the signals and cues necessary for implanted structures to innervate the host, as organs devoid of neural connections provide little benefit to the patient. While developmental studies have identified neuronal pathfinding molecules required for proper patterning during embryogenesis, strategies to initiate innervation in structures transplanted at later times or alternate locations remain limited. Recent work has identified membrane resting potential of nerves as a key regulator of growth cone extension or arrest. Here, we identify a novel role of bioelectricity in the generation of axon guidance cues, showing that neurons read the electric topography of surrounding cells, and demonstrate these cues can be leveraged to initiate sensory organ transplant innervation. Grafts of fluorescently labeled embryological eye primordia were used to produce ectopic eyes in Xenopus laevis tadpoles. Depolarization of host tissues through anion channel activation or other means led to a striking hyperinnervation of the body by these ectopic eyes. A screen of possible transduction mechanisms identified serotonergic signaling to be essential for hyperinnervation to occur, and our molecular data suggest a possible model of bioelectrical control of the distribution of neurotransmitters that guides nerve growth. Together, these results identify the molecular components of bioelectrical signaling among cells that regulates axon guidance, and suggest novel biomedical and bioengineering strategies for triggering neuronal outgrowth using ion channel drugs already approved for human use.
Collapse
Affiliation(s)
- Douglas J. Blackiston
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155 USA
| | - George M. Anderson
- Yale Child Study Center and Department of Laboratory Medicine, Yale University School of Medicine, 230 S. Frontage Rd., New Haven, CT 06519 USA
| | - Nikita Rahman
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155 USA
| | - Clara Bieck
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155 USA
| | - Michael Levin
- Center for Regenerative and Developmental Biology and Department of Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155 USA
| |
Collapse
|
388
|
Oosterhof N, Boddeke E, van Ham TJ. Immune cell dynamics in the CNS: Learning from the zebrafish. Glia 2014; 63:719-35. [PMID: 25557007 DOI: 10.1002/glia.22780] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/10/2014] [Indexed: 12/22/2022]
Abstract
A major question in research on immune responses in the brain is how the timing and nature of these responses influence physiology, pathogenesis or recovery from pathogenic processes. Proper understanding of the immune regulation of the human brain requires a detailed description of the function and activities of the immune cells in the brain. Zebrafish larvae allow long-term, noninvasive imaging inside the brain at high-spatiotemporal resolution using fluorescent transgenic reporters labeling specific cell populations. Together with recent additional technical advances this allows an unprecedented versatility and scope of future studies. Modeling of human physiology and pathology in zebrafish has already yielded relevant insights into cellular dynamics and function that can be translated to the human clinical situation. For instance, in vivo studies in the zebrafish have provided new insight into immune cell dynamics in granuloma formation in tuberculosis and the mechanisms involving treatment resistance. In this review, we highlight recent findings and novel tools paving the way for basic neuroimmunology research in the zebrafish. GLIA 2015;63:719-735.
Collapse
Affiliation(s)
- Nynke Oosterhof
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
389
|
Karlstetter M, Scholz R, Rutar M, Wong WT, Provis JM, Langmann T. Retinal microglia: just bystander or target for therapy? Prog Retin Eye Res 2014; 45:30-57. [PMID: 25476242 DOI: 10.1016/j.preteyeres.2014.11.004] [Citation(s) in RCA: 412] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/20/2014] [Accepted: 11/25/2014] [Indexed: 10/24/2022]
Abstract
Resident microglial cells can be regarded as the immunological watchdogs of the brain and the retina. They are active sensors of their neuronal microenvironment and rapidly respond to various insults with a morphological and functional transformation into reactive phagocytes. There is strong evidence from animal models and in situ analyses of human tissue that microglial reactivity is a common hallmark of various retinal degenerative and inflammatory diseases. These include rare hereditary retinopathies such as retinitis pigmentosa and X-linked juvenile retinoschisis but also comprise more common multifactorial retinal diseases such as age-related macular degeneration, diabetic retinopathy, glaucoma, and uveitis as well as neurological disorders with ocular manifestation. In this review, we describe how microglial function is kept in balance under normal conditions by cross-talk with other retinal cells and summarize how microglia respond to different forms of retinal injury. In addition, we present the concept that microglia play a key role in local regulation of complement in the retina and specify aspects of microglial aging relevant for chronic inflammatory processes in the retina. We conclude that this resident immune cell of the retina cannot be simply regarded as bystander of disease but may instead be a potential therapeutic target to be modulated in the treatment of degenerative and inflammatory diseases of the retina.
Collapse
Affiliation(s)
- Marcus Karlstetter
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Rebecca Scholz
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Matt Rutar
- The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australian Capital Territory, Australia
| | - Wai T Wong
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jan M Provis
- The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australian Capital Territory, Australia
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Cologne, Germany.
| |
Collapse
|
390
|
Ali I, Chugh D, Ekdahl CT. Role of fractalkine-CX3CR1 pathway in seizure-induced microglial activation, neurodegeneration, and neuroblast production in the adult rat brain. Neurobiol Dis 2014; 74:194-203. [PMID: 25461978 DOI: 10.1016/j.nbd.2014.11.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/21/2014] [Accepted: 11/12/2014] [Indexed: 10/24/2022] Open
Abstract
Temporal lobe seizures lead to an acute inflammatory response in the brain primarily characterized by activation of parenchymal microglial cells. Simultaneously, degeneration of pyramidal cells and interneurons is evident together with a seizure-induced increase in the production of new neurons within the dentate gyrus of the hippocampus. We have previously shown a negative correlation between the acute seizure-induced inflammation and the survival of newborn hippocampal neurons. Here, we aimed to evaluate the role of the fractalkine-CX3CR1 pathway for these acute events. Fractalkine is a chemokine expressed by both neurons and glia, while its receptor, CX3CR1 is primarily expressed on microglia. Electrically-induced partial status epilepticus (SE) was induced in adult rats through stereotaxically implanted electrodes in the hippocampus. Recombinant rat fractalkine or CX3CR1 antibody was infused intraventricularly during one week post-SE. A significant increase in the expression of CX3CR1, but not fractalkine, was observed in the dentate gyrus at one week. CX3CR1 antibody treatment resulted in a reduction in microglial activation, neurodegeneration, as well as neuroblast production. In contrast, fractalkine treatment had only minor effects. This study provides evidence for a role of the fractalkine-CX3CR1 signaling pathway in seizure-induced microglial activation and suggests that neuroblast production following seizures may partly occur as a result of microglial activation.
Collapse
Affiliation(s)
- Idrish Ali
- Inflammation and Stem Cell Therapy Group, Wallenberg Neuroscience Center, Division of Clinical Neurophysiology, Lund University, Sweden; Lund Epilepsy Center, Lund University, SE-221 84 Lund, Sweden
| | - Deepti Chugh
- Inflammation and Stem Cell Therapy Group, Wallenberg Neuroscience Center, Division of Clinical Neurophysiology, Lund University, Sweden; Lund Epilepsy Center, Lund University, SE-221 84 Lund, Sweden
| | - Christine T Ekdahl
- Inflammation and Stem Cell Therapy Group, Wallenberg Neuroscience Center, Division of Clinical Neurophysiology, Lund University, Sweden; Lund Epilepsy Center, Lund University, SE-221 84 Lund, Sweden.
| |
Collapse
|
391
|
Abstract
The zebrafish is a premier vertebrate model system that offers many experimental advantages for in vivo imaging and genetic studies. This review provides an overview of glial cell types in the central and peripheral nervous system of zebrafish. We highlight some recent work that exploited the strengths of the zebrafish system to increase the understanding of the role of Gpr126 in Schwann cell myelination and illuminate the mechanisms controlling oligodendrocyte development and myelination. We also summarize similarities and differences between zebrafish radial glia and mammalian astrocytes and consider the possibility that their distinct characteristics may represent extremes in a continuum of cell identity. Finally, we focus on the emergence of zebrafish as a model for elucidating the development and function of microglia. These recent studies have highlighted the power of the zebrafish system for analyzing important aspects of glial development and function.
Collapse
Affiliation(s)
- David A Lyons
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - William S Talbot
- Department of Developmental Biology, Stanford University, Stanford, California 94305
| |
Collapse
|
392
|
Cheung G, Chever O, Rouach N. Connexons and pannexons: newcomers in neurophysiology. Front Cell Neurosci 2014; 8:348. [PMID: 25408635 PMCID: PMC4219455 DOI: 10.3389/fncel.2014.00348] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/06/2014] [Indexed: 11/14/2022] Open
Abstract
Connexin hemichannels are single membrane channels which have been traditionally thought to work in pairs to form gap junction channels across two opposing cells. In astrocytes, gap junction channels allow direct intercellular communication and greatly facilitate the transmission of signals. Recently, there has been growing evidence demonstrating that connexin hemichannels, as well as pannexin channels, on their own are open in various conditions. They allow bidirectional flow of ions and signaling molecules and act as release sites for transmitters like ATP and glutamate into the extracellular space. While much attention has focused on the function of connexin hemichannels and pannexons during pathological situations like epilepsy, inflammation, neurodegeneration or ischemia, their potential roles in physiology is often ignored. In order to fully understand the dynamic properties and roles of connexin hemichannels and pannexons in the brain, it is essential to decipher whether they also have some physiological functions and contribute to normal cerebral processes. Here, we present recent studies in the CNS suggesting emerging physiological functions of connexin hemichannels and pannexons in normal neuronal activity and behavior. We also discuss how these pioneer studies pave the way for future research to extend the physiological relevance of connexons and pannexons, and some fundamental issues yet to be addressed.
Collapse
Affiliation(s)
- Giselle Cheung
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, France
| | - Oana Chever
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, France
| |
Collapse
|
393
|
Kyrargyri V, Vega-Flores G, Gruart A, Delgado-García JM, Probert L. Differential contributions of microglial and neuronal IKKβ to synaptic plasticity and associative learning in alert behaving mice. Glia 2014; 63:549-66. [PMID: 25297800 DOI: 10.1002/glia.22756] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/10/2014] [Accepted: 09/12/2014] [Indexed: 11/11/2022]
Abstract
Microglia are CNS resident immune cells and a rich source of neuroactive mediators, but their contribution to physiological brain processes such as synaptic plasticity, learning, and memory is not fully understood. In this study, we used mice with partial depletion of IκB kinase β, the main activating kinase in the inducible NF-κB pathway, selectively in myeloid lineage cells (mIKKβKO) or excitatory neurons (nIKKβKO) to measure synaptic strength at hippocampal Schaffer collaterals during long-term potentiation (LTP) and instrumental conditioning in alert behaving individuals. Resting microglial cells in mIKKβKO mice showed less Iba1-immunoreactivity, and brain IL-1β mRNA levels were selectively reduced compared with controls. Measurement of field excitatory postsynaptic potentials (fEPSPs) evoked by stimulation of the CA3-CA1 synapse in mIKKβKO mice showed higher facilitation in response to paired pulses and enhanced LTP following high frequency stimulation. In contrast, nIKKβKO mice showed normal basic synaptic transmission and LTP induction but impairments in late LTP. To understand the consequences of such impairments in synaptic plasticity for learning and memory, we measured CA1 fEPSPs in behaving mice during instrumental conditioning. IKKβ was not necessary in either microglia or neurons for mice to learn lever-pressing (appetitive behavior) to obtain food (consummatory behavior) but was required in both for modification of their hippocampus-dependent appetitive, not consummatory behavior. Our results show that microglia, through IKKβ and therefore NF-κB activity, regulate hippocampal synaptic plasticity and that both microglia and neurons, through IKKβ, are necessary for animals to modify hippocampus-driven behavior during associative learning.
Collapse
Affiliation(s)
- Vasiliki Kyrargyri
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, Athens, 11521, Greece
| | | | | | | | | |
Collapse
|
394
|
Neuronal hyperactivity recruits microglial processes via neuronal NMDA receptors and microglial P2Y12 receptors after status epilepticus. J Neurosci 2014; 34:10528-40. [PMID: 25100587 DOI: 10.1523/jneurosci.0416-14.2014] [Citation(s) in RCA: 356] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Microglia are highly dynamic immune cells of the CNS and their dynamism is proposed to be regulated by neuronal activities. However, the mechanisms underlying neuronal regulation of microglial dynamism have not been determined. Here, we found an increased number of microglial primary processes in the hippocampus during KA-induced seizure activity. Consistently, global glutamate induced robust microglial process extension toward neurons in both brain slices and in the intact brain in vivo. The mechanism of the glutamate-induced microglial process extension involves the activation of neuronal NMDA receptors, calcium influx, subsequent ATP release, and microglial response through P2Y12 receptors. Seizure-induced increases in microglial process numbers were also dependent on NMDA receptor activation. Finally, we found that P2Y12 KO mice exhibited reduced seizure-induced increases in microglial process numbers and worsened KA-induced seizure behaviors. Our results elucidate the molecular mechanisms underlying microglia-neuron communication that may be potentially neuroprotective in the epileptic brain.
Collapse
|
395
|
Activation of neuronal NMDA receptors triggers transient ATP-mediated microglial process outgrowth. J Neurosci 2014; 34:10511-27. [PMID: 25100586 DOI: 10.1523/jneurosci.0405-14.2014] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Microglia are morphologically dynamic cells that rapidly extend their processes in response to various stimuli including extracellular ATP. In this study, we tested the hypothesis that stimulation of neuronal NMDARs trigger ATP release leading to communication with microglia. We used acute mouse hippocampal brain slices and two-photon laser scanning microscopy to study microglial dynamics and developed a novel protocol for fixation and immunolabeling of microglia processes. Similar to direct topical ATP application in vivo, short multiple applications of NMDA triggered transient microglia process outgrowth that was reversible and repeatable indicating that this was not due to excitotoxic damage. Stimulation of NMDAR was required as NMDAR antagonists, but not blockers of AMPA/kainate receptors or voltage-gated sodium channels, prevented microglial outgrowth. We report that ATP release, secondary to NMDAR activation, was the key mediator of this neuron-microglia communication as both blocking purinergic receptors and inhibiting hydrolysis of ATP to prevent locally generated gradients abolished outgrowth. Pharmacological and genetic analyses showed that the NMDA-triggered microglia process extension was independent of Pannexin 1, the ATP releasing channels, ATP release from astrocytes via connexins, and nitric oxide generation. Finally, using whole-cell patch clamping we demonstrate that activation of dendritic NMDAR on single neurons is sufficient to trigger microglia process outgrowth. Our results suggest that dendritic neuronal NMDAR activation triggers ATP release via a Pannexin 1-independent manner that induces outgrowth of microglia processes. This represents a novel uncharacterized form of neuron-microglial communication mediated by ATP.
Collapse
|
396
|
Microglia Modulate Wiring of the Embryonic Forebrain. Cell Rep 2014; 8:1271-9. [DOI: 10.1016/j.celrep.2014.07.042] [Citation(s) in RCA: 502] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/28/2014] [Accepted: 07/23/2014] [Indexed: 11/19/2022] Open
|
397
|
Bloom O. Non-mammalian model systems for studying neuro-immune interactions after spinal cord injury. Exp Neurol 2014; 258:130-40. [PMID: 25017894 PMCID: PMC4099969 DOI: 10.1016/j.expneurol.2013.12.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 12/24/2013] [Accepted: 12/26/2013] [Indexed: 01/09/2023]
Abstract
Mammals exhibit poor recovery after injury to the spinal cord, where the loss of neurons and neuronal connections can be functionally devastating. In contrast, it has long been appreciated that many non-mammalian vertebrate species exhibit significant spontaneous functional recovery after spinal cord injury (SCI). Identifying the biological responses that support an organism's inability or ability to recover function after SCI is an important scientific and medical question. While recent advances have been made in understanding the responses to SCI in mammals, we remain without an effective clinical therapy for SCI. A comparative biological approach to understanding responses to SCI in non-mammalian vertebrates will yield important insights into mechanisms that promote recovery after SCI. Presently, mechanistic studies aimed at elucidating responses, both intrinsic and extrinsic to neurons, that result in different regenerative capacities after SCI across vertebrates are just in their early stages. There are several inhibitory mechanisms proposed to impede recovery from SCI in mammals, including reactive gliosis and scarring, myelin associated proteins, and a suboptimal immune response. One hypothesis to explain the robust regenerative capacity of several non-mammalian vertebrates is a lack of some or all of these inhibitory signals. This review presents the current knowledge of immune responses to SCI in several non-mammalian species that achieve anatomical and functional recovery after SCI. This subject is of growing interest, as studies increasingly show both beneficial and detrimental roles of the immune response following SCI in mammals. A long-term goal of biomedical research in all experimental models of SCI is to understand how to promote functional recovery after SCI in humans. Therefore, understanding immune responses to SCI in non-mammalian vertebrates that achieve functional recovery spontaneously may identify novel strategies to modulate immune responses in less regenerative species and promote recovery after SCI.
Collapse
Affiliation(s)
- Ona Bloom
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA; The Hofstra North Shore-LIJ School of Medicine, Hempstead Turnpike, Hempstead, NY 11549, USA.
| |
Collapse
|
398
|
Microglial displacement of inhibitory synapses provides neuroprotection in the adult brain. Nat Commun 2014; 5:4486. [PMID: 25047355 PMCID: PMC4109015 DOI: 10.1038/ncomms5486] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 06/23/2014] [Indexed: 01/21/2023] Open
Abstract
Microglia actively survey the brain microenvironment and play essential roles in sculpting synaptic connections during brain development. While microglial functions in the adult brain are less clear, activated microglia can closely appose neuronal cell bodies and displace axosomatic presynaptic terminals. Microglia-mediated stripping of presynaptic terminals is considered neuroprotective, but the cellular and molecular mechanisms are poorly defined. Using 3D electron microscopy, we demonstrate that activated microglia displace inhibitory presynaptic terminals from cortical neurons in adult mice. Electrophysiological recordings further establish that the reduction in inhibitory GABAergic synapses increased synchronized firing of cortical neurons in γ-frequency band. Increased neuronal activity results in the calcium-mediated activation of CaM kinase IV, phosphorylation of CREB, increased expression of antiapoptotic and neurotrophic molecules and reduced apoptosis of cortical neurons following injury. These results indicate that activated microglia can protect the adult brain by migrating to inhibitory synapses and displacing them from cortical neurons. Microglia play essential roles in sculpting synaptic connections during brain development but their role in the adult brain is less clear. Here the authors show that activated microglia can prophylactically protect the adult rodent brain from injury by migrating to and displacing inhibitory synapses from cortical neurons.
Collapse
|
399
|
Santiago AR, Baptista FI, Santos PF, Cristóvão G, Ambrósio AF, Cunha RA, Gomes CA. Role of microglia adenosine A(2A) receptors in retinal and brain neurodegenerative diseases. Mediators Inflamm 2014; 2014:465694. [PMID: 25132733 PMCID: PMC4124703 DOI: 10.1155/2014/465694] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/20/2014] [Indexed: 12/20/2022] Open
Abstract
Neuroinflammation mediated by microglial cells in the brain has been commonly associated with neurodegenerative diseases. Whether this microglia-mediated neuroinflammation is cause or consequence of neurodegeneration is still a matter of controversy. However, it is unequivocal that chronic neuroinflammation plays a role in disease progression and halting that process represents a potential therapeutic strategy. The neuromodulator adenosine emerges as a promising targeting candidate based on its ability to regulate microglial proliferation, chemotaxis, and reactivity through the activation of its G protein coupled A2A receptor (A2AR). This is in striking agreement with the ability of A2AR blockade to control several brain diseases. Retinal degenerative diseases have been also associated with microglia-mediated neuroinflammation, but the role of A2AR has been scarcely explored. This review aims to compare inflammatory features of Parkinson's and Alzheimer's diseases with glaucoma and diabetic retinopathy, discussing the therapeutic potential of A2AR in these degenerative conditions.
Collapse
Affiliation(s)
- Ana R. Santiago
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- AIBILI, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Largo Marquês de Pombal, Universidade de Coimbra, 3004-517 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Filipa I. Baptista
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Paulo F. Santos
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Largo Marquês de Pombal, Universidade de Coimbra, 3004-517 Coimbra, Portugal
- Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Gonçalo Cristóvão
- Center for Neuroscience and Cell Biology, Largo Marquês de Pombal, Universidade de Coimbra, 3004-517 Coimbra, Portugal
| | - António F. Ambrósio
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- AIBILI, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Largo Marquês de Pombal, Universidade de Coimbra, 3004-517 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Rodrigo A. Cunha
- Center for Neuroscience and Cell Biology, Largo Marquês de Pombal, Universidade de Coimbra, 3004-517 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| | - Catarina A. Gomes
- Centre of Ophthalmology and Vision Sciences, IBILI, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology, Largo Marquês de Pombal, Universidade de Coimbra, 3004-517 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| |
Collapse
|
400
|
Salter M, Beggs S. Sublime Microglia: Expanding Roles for the Guardians of the CNS. Cell 2014; 158:15-24. [DOI: 10.1016/j.cell.2014.06.008] [Citation(s) in RCA: 422] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Indexed: 12/31/2022]
|