101
|
Embryonic Neocortical Microglia Express Toll-Like Receptor 9 and Respond to Plasmid DNA Injected into the Ventricle: Technical Considerations Regarding Microglial Distribution in Electroporated Brain Walls. eNeuro 2018; 5:eN-MNT-0312-18. [PMID: 30627652 PMCID: PMC6325556 DOI: 10.1523/eneuro.0312-18.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/19/2018] [Accepted: 10/27/2018] [Indexed: 12/25/2022] Open
Abstract
Microglia, the resident immune cells in the CNS, play multiple roles during development. In the embryonic cerebral wall, microglia modulate the functions of neural stem/progenitor cells through their distribution in regions undergoing cell proliferation and/or differentiation. Previous studies using CX3CR1-GFP transgenic mice demonstrated that microglia extensively survey these regions. To simultaneously visualize microglia and neural-lineage cells that interact with each other, we applied the in utero electroporation (IUE) technique, which has been widely used for gene-transfer in neurodevelopmental studies, to CX3CR1-GFP mice (males and females). However, we unexpectedly faced a technical problem: although microglia are normally distributed homogeneously throughout the mid-embryonic cortical wall with only limited luminal entry, the intraventricular presence of exogenously derived plasmid DNAs induced microglia to accumulate along the apical surface of the cortex and aggregate in the choroid plexus. This effect was independent of capillary needle puncture of the brain wall or application of electrical pulses. The microglial response occurred at plasmid DNA concentrations lower than those routinely used for IUE, and was mediated by activation of Toll-like receptor 9 (TLR9), an innate immune sensor that recognizes unmethylated cytosine-phosphate guanosine motifs abundant in microbial DNA. Administration of plasmid DNA together with oligonucleotide 2088, the antagonist of TLR9, partially restored the dispersed intramural localization of microglia and significantly decreased luminal accumulation of these cells. Thus, via TLR9, intraventricular plasmid DNA administration causes aberrant distribution of embryonic microglia, suggesting that the behavior of microglia in brain primordia subjected to IUE should be carefully interpreted.
Collapse
|
102
|
Abstract
Cross-talk between the nervous and immune systems has been well described in the context of adult physiology and disease. Recent advances in our understanding of immune cell ontogeny have revealed a notable interplay between neurons and microglia during the prenatal and postnatal emergence of functional circuits. This Review focuses on the brain, where the early symbiotic relationship between microglia and neuronal cells critically regulates wiring, contributes to sex-specific differences in neural circuits, and relays crucial information from the periphery, including signals derived from the microbiota. These observations underscore the importance of studying neurodevelopment as part of a broader framework that considers nervous system interactions with microglia in a whole-body context.
Collapse
|
103
|
Smolders S, Notter T, Smolders SMT, Rigo JM, Brône B. Controversies and prospects about microglia in maternal immune activation models for neurodevelopmental disorders. Brain Behav Immun 2018; 73:51-65. [PMID: 29870753 DOI: 10.1016/j.bbi.2018.06.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/26/2018] [Accepted: 06/01/2018] [Indexed: 12/16/2022] Open
Abstract
Activation of the maternal immune system during pregnancy is a well-established risk factor for neuropsychiatric disease in the offspring, yet, the underlying mechanisms leading to altered brain function remain largely undefined. Microglia, the resident immune cells of the brain, are key to adequate development of the central nervous system (CNS), and are prime candidates to mediate maternal immune activation (MIA)-induced brain abnormalities. As such, the effects of MIA on the immunological phenotype of microglia has been widely investigated. However, contradicting results due to differences in read-out and methodological approaches impede final conclusions on MIA-induced microglial alterations. The aim of this review is to critically discuss the evidence for an activated microglial phenotype upon MIA.
Collapse
Affiliation(s)
- Silke Smolders
- Uhasselt - BIOMED, Hasselt, Belgium; Laboratory of Neuronal Differentiation, VIB Center for the Biology of Disease, Leuven and Center for Human Genetics, KU Leuven Leuven, Belgium.
| | - Tina Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| | - Sophie M T Smolders
- Uhasselt - BIOMED, Hasselt, Belgium; INSERM, UMR S 1130, Université Pierre et Marie Curie Paris, France; CNRS, UMR 8246, Université Pierre et Marie Curie Paris, France; UM 119 NPS, Université Pierre et Marie Curie Paris, France.
| | | | | |
Collapse
|
104
|
Angelim MKSC, Maia LMSDS, Mouffle C, Ginhoux F, Low D, Amancio-Dos-Santos A, Makhoul J, Le Corronc H, Mangin JM, Legendre P. Embryonic macrophages and microglia ablation alter the development of dorsal root ganglion sensory neurons in mouse embryos. Glia 2018; 66:2470-2486. [PMID: 30252950 DOI: 10.1002/glia.23499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/29/2018] [Accepted: 07/03/2018] [Indexed: 12/15/2022]
Abstract
Microglia are known to regulate several aspects of the development of the central nervous system. When microglia colonize the spinal cord, from E11.5 in the mouse embryo, they interact with growing central axons of dorsal root ganglion sensory neurons (SNs), which suggests that they may have some functions in SN development. To address this issue, we analyzed the effects of embryonic macrophage ablation on the early development of SNs using mouse embryo lacking embryonic macrophages (PU.1 knock-out mice) and immune cell ablation. We discovered that, in addition to microglia, embryonic macrophages contact tropomyosin receptor kinase (Trk) C+ SN, TrkB+ SN, and TrkA+ SN peripheral neurites from E11.5. Deprivation of immune cells resulted in an initial reduction of TrkC+ SN and TrkB+ SN populations at E11.5 that was unlikely to be related to an alteration in their developmental cell death (DCD), followed by a transitory increase in their number at E12.5. It also resulted in a reduction of TrkA+ SN number during the developmental period analyzed (E11.5-E15.5), although we did not observe any change in their DCD. Proliferation of cells negative for brain fatty acid-binding protein (BFABP- ), which likely correspond to neuronal progenitors, was increased at E11.5, while their proliferation was decreased at E12.5, which could partly explain the alterations of SN subtype production observed from E11.5. In addition, we observed alterations in the proliferation of glial cell progenitors (BFABP+ cells) in the absence of embryonic macrophages. Our data indicate that embryonic macrophages and microglia ablation alter the development of SNs.
Collapse
Affiliation(s)
- Monara Kaélle Sérvulo Cruz Angelim
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France.,Neurophysiology and pharmacology laboratory, Federal University of Pernambuco, Pernambuco, Brazil
| | - Luciana Maria Silva de Seixas Maia
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France.,Neurophysiology and pharmacology laboratory, Federal University of Pernambuco, Pernambuco, Brazil
| | - Christine Mouffle
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | | | - Jennifer Makhoul
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France
| | - Hervé Le Corronc
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France.,Université d'Angers, Angers, France
| | - Jean-Marie Mangin
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France
| | - Pascal Legendre
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neurosciences Paris Seine, Institut de Biologie Paris Seine (NPS, IBPS), Paris, France
| |
Collapse
|
105
|
Abstract
Microglia, the primary resident immune cell type, constitute a key population of glia in the retina. Recent evidence indicates that microglia play significant functional roles in the retina at different life stages. During development, retinal microglia regulate neuronal survival by exerting trophic influences and influencing programmed cell death. During adulthood, ramified microglia in the plexiform layers interact closely with synapses to maintain synaptic structure and function that underlie the retina's electrophysiological response to light. Under pathological conditions, retinal microglia participate in potentiating neurodegeneration in diseases such as glaucoma, retinitis pigmentosa, and age-related neurodegeneration by producing proinflammatory neurotoxic cytokines and removing living neurons via phagocytosis. Modulation of pathogenic microglial activation states and effector mechanisms has been linked to neuroprotection in animal models of retinal diseases. These findings have led to the design of early proof-of-concept clinical trials with microglial modulation as a therapeutic strategy.
Collapse
Affiliation(s)
- Sean M. Silverman
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;,
| | - Wai T. Wong
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;,
| |
Collapse
|
106
|
Silvin A, Ginhoux F. Microglia heterogeneity along a spatio-temporal axis: More questions than answers. Glia 2018; 66:2045-2057. [PMID: 30144321 DOI: 10.1002/glia.23458] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/05/2018] [Accepted: 05/04/2018] [Indexed: 12/29/2022]
Abstract
Microglia are resident macrophages of the central nervous system; they arise during early embryonic development and persist throughout adulthood. These unique cells provide developmental support, contribute to adult brain homeostasis and impart immune protection during infection. Dysregulated microglia are implicated in the pathophysiology of several neurological disorders, including Alzheimer disease, and as such, a better understanding of their regulation and function is required for rational therapeutic design. Recent studies have highlighted the various heterogeneous aspects of microglia, such as their wide differentiation spectrum from early embryogenesis to adulthood, their location in different brain regions and their responses to ageing, infection and inflammation. In this review, we discuss microglial heterogeneity in time and space and highlight the remaining questions arising from such heterogeneity.
Collapse
Affiliation(s)
- Aymeric Silvin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, IMMUNOS Building #3-4, BIOPOLIS, 138648, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, IMMUNOS Building #3-4, BIOPOLIS, 138648, Singapore.,Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
107
|
Hattori Y, Miyata T. Microglia extensively survey the developing cortex via the CXCL12/CXCR4 system to help neural progenitors to acquire differentiated properties. Genes Cells 2018; 23:915-922. [PMID: 30144249 DOI: 10.1111/gtc.12632] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 02/04/2023]
Abstract
Neocortical development proceeds through the formation of new zones in which neural-lineage cells are organized based on their differentiation status. Although microglia initially distribute homogeneously throughout the growing cerebral wall, they accumulate in the inner cytogenic zone, the ventricular zone (VZ) and the subventricular zone (SVZ) in the mid-embryonic stage. However, the roles of these cells remain to be elucidated. In this study, we found that microglia, despite being only a minor population of the cells that constitute the cerebral wall, promote the differentiation of neural progenitor cells by frequently moving throughout the cortex; their migration is mediated by the CXCL12/CXCR4 system. Pulse-chase experiments confirmed that microglia help Pax6+ stem-like cells to differentiate into Tbr2+ intermediate progenitors. Further, monitoring of microglia by live imaging showed that administration of AMD3100, an antagonist of CXCR4, dampened microglial movement and decreased microglial surveillance throughout the cortex. In particular, arrest of microglial motion led to a prominent decrease in the abundance of Tbr2+ cells in the SVZ. Based on our findings, we propose that extensive surveillance by microglia contributes to the efficient functioning of these cells, thereby regulating the differentiation of neural stem-like cells.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
108
|
Fehrenbach M, Tjwa M, Bechmann I, Krueger M. Decreased microglial numbers in Vav1-Cre + :dicer knock-out mice suggest a second source of microglia beyond yolk sac macrophages. Ann Anat 2018; 218:190-198. [DOI: 10.1016/j.aanat.2018.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 01/05/2023]
|
109
|
Distribution and Morphological Features of Microglia in the Developing Cerebral Cortex of Gyrencephalic Mammals. Neurochem Res 2018; 43:1075-1085. [PMID: 29616442 DOI: 10.1007/s11064-018-2520-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 02/23/2018] [Accepted: 03/27/2018] [Indexed: 12/11/2022]
Abstract
Microglia have been attracting much attention because of their fundamental importance in both the mature brain and the developing brain. Though important roles of microglia in the developing cerebral cortex of mice have been uncovered, their distribution and roles in the developing cerebral cortex in gyrencephalic higher mammals have remained elusive. Here we examined the distribution and morphology of microglia in the developing cerebral cortex of gyrencephalic carnivore ferrets. We found that a number of microglia were accumulated in the germinal zones (GZs), especially in the outer subventricular zone (OSVZ), which is a GZ found in higher mammals. Furthermore, we uncovered that microglia extended their processes tangentially along inner fiber layer (IFL)-like fibers in the developing ferret cortex. The OSVZ and the IFL are the prominent features of the cerebral cortex of higher mammals. Our findings indicate that microglia may play important roles in the OSVZ and the IFL in the developing cerebral cortex of higher mammals.
Collapse
|
110
|
Bittle J, Stevens HE. The role of glucocorticoid, interleukin-1β, and antioxidants in prenatal stress effects on embryonic microglia. J Neuroinflammation 2018; 15:44. [PMID: 29452586 PMCID: PMC5815231 DOI: 10.1186/s12974-018-1079-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/24/2018] [Indexed: 12/24/2022] Open
Abstract
Maternal stress during pregnancy is associated with an increased risk of psychopathology in offspring. Resident immune cells of the brain, microglia, may be mediators of prenatal stress and altered neurodevelopment. Here, we demonstrate that neither the exogenous pro-inflammatory cytokine, interleukin-1β (IL-1β), nor the glucocorticoid hormone, corticosterone, recapitulated the full effects of prenatal stress on the morphology of microglial cells in the cortical plate of embryonic mice; IL-1β effects showed greater similarity to prenatal stress effects on microglia. Unexpectedly, oil vehicle alone, which has antioxidant properties, moderated the effects of prenatal stress on microglia. Microglia changes with prenatal stress were also sensitive to the antioxidant, N-acetylcysteine, suggesting redox dysregulation as a mechanism of prenatal stress.
Collapse
Affiliation(s)
- Jada Bittle
- Department of Psychiatry, University of Iowa Carver College of Medicine, 1330 PBDB, 169 Newton Rd., Iowa City, IA 52246 USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, 356 Medical Research Center, Iowa City, IA 52242 USA
| | - Hanna E. Stevens
- Department of Psychiatry, University of Iowa Carver College of Medicine, 1330 PBDB, 169 Newton Rd., Iowa City, IA 52246 USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, 356 Medical Research Center, Iowa City, IA 52242 USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, 2312 PBDB, 169 Newton Rd., Iowa City, IA 52246 USA
| |
Collapse
|
111
|
Interleukin 4 modulates microglia homeostasis and attenuates the early slowly progressive phase of amyotrophic lateral sclerosis. Cell Death Dis 2018; 9:250. [PMID: 29445154 PMCID: PMC5833860 DOI: 10.1038/s41419-018-0288-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/28/2017] [Accepted: 01/04/2018] [Indexed: 12/31/2022]
Abstract
Microglia activation is a commonly pathological hallmark of neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), a devastating disorder characterized by a selective motor neurons degeneration. Whether such activation might represent a causal event rather than a secondary epiphenomenon remains elusive. Here, we show that CNS-delivery of IL-4—via a lentiviral-mediated gene therapy strategy—skews microglia to proliferate, inducing these cells to adopt the phenotype of slowly proliferating cells. Transcriptome analysis revealed that IL-4-treated microglia express a broad number of genes normally encoded by embryonic microglia. Since embryonic microglia sustain CNS development, we then hypothesized that turning adult microglia to acquire such phenotype via IL-4 might be an efficient in vivo strategy to sustain motor neuron survival in ALS. IL-4 gene therapy in SOD1G93A mice resulted in a general amelioration of clinical outcomes during the early slowly progressive phase of the disease. However, such approach did not revert neurodegenerative processes occurring in the late and fast progressing phase of the disease.
Collapse
|
112
|
Low D, Ginhoux F. Recent advances in the understanding of microglial development and homeostasis. Cell Immunol 2018; 330:68-78. [PMID: 29366562 DOI: 10.1016/j.cellimm.2018.01.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/06/2018] [Accepted: 01/07/2018] [Indexed: 01/04/2023]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS). These pivotal cells arise early during embryonic development and provide both developmental support and immune protection to the brain. In adults, microglia contribute to brain homeostasis and mediate an intriguing interplay between the CNS and the gut microbiota. When dysregulated, microglia are also implicated in numerous neurological disorders, and thus fully understanding their regulation and functions will facilitate rational design of therapies to alleviate these conditions; however it remains unclear how the multiple factors modulating microglial activity are integrated at the organism and cellular levels. In this review, we will discuss recent advances in the understanding of microglial regulation and highlight the key questions that remain to be answered around microglial development, homeostasis and functions.
Collapse
Affiliation(s)
- Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore; Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
113
|
Tay TL, Béchade C, D'Andrea I, St-Pierre MK, Henry MS, Roumier A, Tremblay ME. Microglia Gone Rogue: Impacts on Psychiatric Disorders across the Lifespan. Front Mol Neurosci 2018; 10:421. [PMID: 29354029 PMCID: PMC5758507 DOI: 10.3389/fnmol.2017.00421] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022] Open
Abstract
Microglia are the predominant immune response cells and professional phagocytes of the central nervous system (CNS) that have been shown to be important for brain development and homeostasis. These cells present a broad spectrum of phenotypes across stages of the lifespan and especially in CNS diseases. Their prevalence in all neurological pathologies makes it pertinent to reexamine their distinct roles during steady-state and disease conditions. A major question in the field is determining whether the clustering and phenotypical transformation of microglial cells are leading causes of pathogenesis, or potentially neuroprotective responses to the onset of disease. The recent explosive growth in our understanding of the origin and homeostasis of microglia, uncovering their roles in shaping of the neural circuitry and synaptic plasticity, allows us to discuss their emerging functions in the contexts of cognitive control and psychiatric disorders. The distinct mesodermal origin and genetic signature of microglia in contrast to other neuroglial cells also make them an interesting target for the development of therapeutics. Here, we review the physiological roles of microglia, their contribution to the effects of environmental risk factors (e.g., maternal infection, early-life stress, dietary imbalance), and their impact on psychiatric disorders initiated during development (e.g., Nasu-Hakola disease (NHD), hereditary diffuse leukoencephaly with spheroids, Rett syndrome, autism spectrum disorders (ASDs), and obsessive-compulsive disorder (OCD)) or adulthood (e.g., alcohol and drug abuse, major depressive disorder (MDD), bipolar disorder (BD), schizophrenia, eating disorders and sleep disorders). Furthermore, we discuss the changes in microglial functions in the context of cognitive aging, and review their implication in neurodegenerative diseases of the aged adult (e.g., Alzheimer’s and Parkinson’s). Taking into account the recent identification of microglia-specific markers, and the availability of compounds that target these cells selectively in vivo, we consider the prospect of disease intervention via the microglial route.
Collapse
Affiliation(s)
- Tuan Leng Tay
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Catherine Béchade
- INSERM UMR-S 839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Ivana D'Andrea
- INSERM UMR-S 839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris, France.,Institut du Fer à Moulin, Paris, France
| | | | - Mathilde S Henry
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Anne Roumier
- INSERM UMR-S 839, Paris, France.,Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Marie-Eve Tremblay
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada.,Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
| |
Collapse
|
114
|
VanRyzin JW, Pickett LA, McCarthy MM. Microglia: Driving critical periods and sexual differentiation of the brain. Dev Neurobiol 2018; 78:580-592. [PMID: 29243403 DOI: 10.1002/dneu.22569] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 12/05/2017] [Accepted: 12/13/2017] [Indexed: 12/12/2022]
Abstract
The proverbial role of microglia during brain development is shifting from passive members of the brain's immune system to active participants that are able to dictate enduring outcomes. Despite these advances, little attention has been paid to one of the most critical components of early brain development-sexual differentiation. Mounting evidence suggests that the normal developmental functions microglia perform-cell number regulation and synaptic connectivity-may be involved in the sex-specific patterning of the brain during these early sensitive periods, and may have lasting sex-dependent and sex-independent effects on behavior. In this review, we outline the known functions of microglia during developmental sensitive periods, and highlight the role they play in the establishment of sex differences in brain and behavior. We also propose a framework for how researchers can incorporate microglia in their study of sex differences and vice versa. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 580-592, 2018.
Collapse
Affiliation(s)
- Jonathan W VanRyzin
- Department of Pharmacology, The University of Maryland School of Medicine, Baltimore, Maryland, 21201.,Program in Neuroscience, The University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Lindsay A Pickett
- Department of Pharmacology, The University of Maryland School of Medicine, Baltimore, Maryland, 21201.,Program in Neuroscience, The University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Margaret M McCarthy
- Department of Pharmacology, The University of Maryland School of Medicine, Baltimore, Maryland, 21201.,Program in Neuroscience, The University of Maryland School of Medicine, Baltimore, Maryland, 21201
| |
Collapse
|
115
|
Weinhard L, d'Errico P, Leng Tay T. Headmasters: Microglial regulation of learning and memory in health and disease. AIMS MOLECULAR SCIENCE 2018. [DOI: 10.3934/molsci.2018.1.63] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
116
|
Burke NN, Fan CY, Trang T. Microglia in health and pain: impact of noxious early life events. Exp Physiol 2018; 101:1003-21. [PMID: 27474262 DOI: 10.1113/ep085714] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/27/2016] [Indexed: 01/08/2023]
Abstract
NEW FINDINGS What is the topic of this review? This review discusses the origins and development of microglia, and how stress, pain or inflammation in early life disturbs microglial function during critical developmental periods, leading to altered pain sensitivity and/or increased risk of chronic pain in later life. What advances does it highlight? We highlight recent advances in understanding how disrupted microglial function impacts the developing nervous system and the consequences for pain processing and susceptibility for development of chronic pain in later life. The discovery of microglia is accredited to Pío del Río-Hortega, who recognized this 'third element' of CNS cells as being morphologically distinct from neurons and astrocytes. For decades after this finding, microglia were altogether ignored or relegated as simply being support cells. Emerging from virtual obscurity, microglia have now gained notoriety as immune cells that assume a leading role in the development, maintenance and protection of a healthy CNS. Pioneering studies have recently shed light on the origins of microglia, their role in the developing nervous system and the complex roles they play beyond the immune response. These studies reveal that altered microglial function can have a profoundly negative impact on the developing brain and may be a determinant in a range of neurodevelopmental disorders and neurodegenerative diseases. The realization that aberrant microglial function also critically underlies chronic pain, a debilitating disorder that afflicts over 1.5 billion people worldwide, was a major conceptual leap forward in the pain field. Adding to this advance is emerging evidence that early life noxious experiences can have a long-lasting impact on central pain processing and adult pain sensitivity. With microglia now coming of age, in this review we examine the association between adverse early life events, such as stress, injury or inflammation, and the influence of sex differences, on the role of microglia in pain physiology in adulthood.
Collapse
Affiliation(s)
- Nikita N Burke
- Department of Comparative Biology and Experimental Medicine, Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Churmy Y Fan
- Department of Comparative Biology and Experimental Medicine, Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Tuan Trang
- Department of Comparative Biology and Experimental Medicine, Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
117
|
Abstract
Although autism spectrum disorder (ASD) has a strong genetic basis, its etiology is complex, with several genetic factors likely to be involved as well as environmental factors. Immune dysregulation has gained significant attention as a causal mechanism in ASD pathogenesis. ASD has been associated with immune abnormalities in the brain and periphery, including inflammatory disorders and autoimmunity in not only the affected individuals but also their mothers. Prenatal exposure to maternal immune activation (MIA) has been implicated as an environmental risk factor for ASD. In support of this notion, animal models have shown that MIA results in offspring with behavioral, neurological, and immunological abnormalities similar to those observed in ASD. This raises the question of how MIA exposure can lead to ASD in susceptible individuals. Recent evidence points to a potential inflammation pathway linking MIA-associated ASD with the activity of T helper 17 (Th17) lymphocytes and their effector cytokine interleukin-17A (IL-17A). IL-17A has been implicated from human studies and elevated IL-17A levels in the blood have been found to correlate with phenotypic severity in a subset of ASD individuals. In MIA model mice, elevated IL-17A levels also have been observed. Additionally, antibody blockade to inhibit IL-17A signaling was found to prevent ASD-like behaviors in offspring exposed to MIA. Therefore, IL-17A dysregulation may play a causal role in the development of ASD. The source of increased IL-17A in the MIA mouse model was attributed to maternal Th17 cells because genetic removal of the transcription factor RORγt to selectively inhibit Th17 differentiation in pregnant mice was able to prevent ASD-like behaviors in the offspring. Similar to ASD individuals, the MIA-exposed offspring also displayed cortical dysplasia which could be prevented by inhibition of IL-17A signaling in pregnant mice. This finding reveals one possible cellular mechanism through which ASD-related cognitive and behavioral deficits may emerge following maternal inflammation. IL-17A can exert strong effects on cell survival and differentiation and the activity of signal transduction cascades, which can have important consequences during cortical development on neural function. This review examines IL-17A signaling pathways in the context of both immunity and neural function that may contribute to the development of ASD associated with MIA.
Collapse
Affiliation(s)
- Helen Wong
- Institute for Behavioral Genetics, University of Colorado-Boulder, CO 80303, United States; Department of Integrative Physiology, University of Colorado-Boulder, Boulder, CO 80303, United States; Linda Crnic Institute, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Charles Hoeffer
- Institute for Behavioral Genetics, University of Colorado-Boulder, CO 80303, United States; Department of Integrative Physiology, University of Colorado-Boulder, Boulder, CO 80303, United States; Linda Crnic Institute, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
118
|
Morton MC, Neckles VN, Feliciano DM. Isolation of Extracellular Vesicles from Subventricular Zone Neural Stem Cells. Methods Mol Biol 2018; 2002:75-85. [PMID: 30244437 DOI: 10.1007/7651_2018_183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The neonatal subventricular zone (SVZ) is a neurogenic niche that contains neural stem cells (NSCs). NSCs release particles called extracellular vesicles (EVs) that contain biological material. EVs are transferred to cells, including immune cells in the brain called microglia. A standard approach to identify EV functions is to isolate and transplant EVs. Here, a detailed protocol is provided that will allow one to culture neonatal SVZ NSCs and to isolate, label, and transplant EVs. The protocol will permit careful and thorough examination of EVs in a wide range of physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Mary C Morton
- Department of Biological Sciences, Clemson University, Clemson, SC, USA
| | | | - David M Feliciano
- Department of Biological Sciences, Clemson University, Clemson, SC, USA.
| |
Collapse
|
119
|
Thion MS, Low D, Silvin A, Chen J, Grisel P, Schulte-Schrepping J, Blecher R, Ulas T, Squarzoni P, Hoeffel G, Coulpier F, Siopi E, David FS, Scholz C, Shihui F, Lum J, Amoyo AA, Larbi A, Poidinger M, Buttgereit A, Lledo PM, Greter M, Chan JKY, Amit I, Beyer M, Schultze JL, Schlitzer A, Pettersson S, Ginhoux F, Garel S. Microbiome Influences Prenatal and Adult Microglia in a Sex-Specific Manner. Cell 2017; 172:500-516.e16. [PMID: 29275859 PMCID: PMC5786503 DOI: 10.1016/j.cell.2017.11.042] [Citation(s) in RCA: 526] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 11/15/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023]
Abstract
Microglia are embryonically seeded macrophages that contribute to brain development, homeostasis, and pathologies. It is thus essential to decipher how microglial properties are temporally regulated by intrinsic and extrinsic factors, such as sexual identity and the microbiome. Here, we found that microglia undergo differentiation phases, discernable by transcriptomic signatures and chromatin accessibility landscapes, which can diverge in adult males and females. Remarkably, the absence of microbiome in germ-free mice had a time and sexually dimorphic impact both prenatally and postnatally: microglia were more profoundly perturbed in male embryos and female adults. Antibiotic treatment of adult mice triggered sexually biased microglial responses revealing both acute and long-term effects of microbiota depletion. Finally, human fetal microglia exhibited significant overlap with the murine transcriptomic signature. Our study shows that microglia respond to environmental challenges in a sex- and time-dependent manner from prenatal stages, with major implications for our understanding of microglial contributions to health and disease. Microglia undergo sequential phases of differentiation during development The maternal microbiome influences microglial properties during prenatal stages The absence of the microbiome has a sex- and time-specific impact on microglia Microbiome depletions have acute and long-term effects on microglial properties
Collapse
Affiliation(s)
- Morgane Sonia Thion
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Aymeric Silvin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Pauline Grisel
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Jonas Schulte-Schrepping
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Ronnie Blecher
- Department of Immunology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Thomas Ulas
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Paola Squarzoni
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Guillaume Hoeffel
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille, France
| | - Fanny Coulpier
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Eleni Siopi
- Institut Pasteur, Unité Perception et Mémoire, CNRS, UMR 3571, F-75015 Paris, France
| | - Friederike Sophie David
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Claus Scholz
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Foo Shihui
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Josephine Lum
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | | | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Anne Buttgereit
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Pierre-Marie Lledo
- Institut Pasteur, Unité Perception et Mémoire, CNRS, UMR 3571, F-75015 Paris, France
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore; KK Research Centre, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore 229899, Singapore
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Marc Beyer
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany; Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Joachim Ludwig Schultze
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany; Platform of Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, 53175 Bonn, Germany
| | - Andreas Schlitzer
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Myeloid Cell Biology, LIMES-Institute, University of Bonn, 53115 Bonn, Germany
| | - Sven Pettersson
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore; Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17165, Sweden
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore.
| | - Sonia Garel
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France.
| |
Collapse
|
120
|
Lannes N, Eppler E, Etemad S, Yotovski P, Filgueira L. Microglia at center stage: a comprehensive review about the versatile and unique residential macrophages of the central nervous system. Oncotarget 2017; 8:114393-114413. [PMID: 29371994 PMCID: PMC5768411 DOI: 10.18632/oncotarget.23106] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/15/2017] [Indexed: 02/07/2023] Open
Abstract
Microglia cells are the unique residential macrophages of the central nervous system (CNS). They have a special origin, as they derive from the embryonic yolk sac and enter the developing CNS at a very early stage. They play an important role during CNS development and adult homeostasis. They have a major contribution to adult neurogenesis and neuroinflammation. Thus, they participate in the pathogenesis of neurodegenerative diseases and contribute to aging. They play an important role in sustaining and breaking the blood-brain barrier. As innate immune cells, they contribute substantially to the immune response against infectious agents affecting the CNS. They play also a major role in the growth of tumours of the CNS. Microglia are consequently the key cell population linking the nervous and the immune system. This review covers all different aspects of microglia biology and pathology in a comprehensive way.
Collapse
Affiliation(s)
- Nils Lannes
- Albert Gockel, Anatomy, Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Elisabeth Eppler
- Pestalozzistrasse Zo, Department of BioMedicine, University of Basel, CH-4056 Basel, Switzerland
| | - Samar Etemad
- Building 71/218 RBWH Herston, Centre for Clinical Research, The University of Queensland, QLD 4029 Brisbane, Australia
| | - Peter Yotovski
- Albert Gockel, Anatomy, Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| | - Luis Filgueira
- Albert Gockel, Anatomy, Department of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| |
Collapse
|
121
|
Sominsky L, De Luca S, Spencer SJ. Microglia: Key players in neurodevelopment and neuronal plasticity. Int J Biochem Cell Biol 2017; 94:56-60. [PMID: 29197626 DOI: 10.1016/j.biocel.2017.11.012] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/15/2017] [Accepted: 11/27/2017] [Indexed: 10/18/2022]
Abstract
Microglia are the primary innate immune cells in the CNS. Since their initial discovery and characterization, decades of research have revealed their unique roles not only in maintaining immune homeostasis, but also being indispensable to brain development and cognitive function. As such, microglia drive synaptogenesis, synaptic pruning, neurogenesis and neuronal activity. Microglia-specific mutations are implicated in several neurodevelopmental disorders, and dysregulation of microglial function is strongly linked to several pathologies, including cognitive decline and Alzheimer's disease. Importantly, developmental insults can lead to long-term changes in microglial function that may compromise the ability of the adult brain to fight infections and process cognitive information. Adult lifestyle or injury can also lastingly influence microglial morphology and function. Here we highlight key research on microglia's role in neuronal plasticity across the lifespan.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia
| | - Simone De Luca
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Vic., 3083, Australia.
| |
Collapse
|
122
|
Neuregulin 3 Mediates Cortical Plate Invasion and Laminar Allocation of GABAergic Interneurons. Cell Rep 2017; 18:1157-1170. [PMID: 28147272 PMCID: PMC5300889 DOI: 10.1016/j.celrep.2016.12.089] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/08/2016] [Accepted: 12/27/2016] [Indexed: 02/07/2023] Open
Abstract
Neural circuits in the cerebral cortex consist of excitatory pyramidal cells and inhibitory interneurons. These two main classes of cortical neurons follow largely different genetic programs, yet they assemble into highly specialized circuits during development following a very precise choreography. Previous studies have shown that signals produced by pyramidal cells influence the migration of cortical interneurons, but the molecular nature of these factors has remained elusive. Here, we identified Neuregulin 3 (Nrg3) as a chemoattractive factor expressed by developing pyramidal cells that guides the allocation of cortical interneurons in the developing cortical plate. Gain- and loss-of-function approaches reveal that Nrg3 modulates the migration of interneurons into the cortical plate in a process that is dependent on the tyrosine kinase receptor ErbB4. Perturbation of Nrg3 signaling in conditional mutants leads to abnormal lamination of cortical interneurons. Nrg3 is therefore a critical mediator in the assembly of cortical inhibitory circuits. Nrg3 acts a short-range chemoattractive molecule for cortical interneurons Nrg3 functions through ErbB4 to attract interneurons into the cortical plate Interneurons prefer Cxcl12 over Nrg3 during tangential migration Disruption of Nrg3 signaling causes abnormal interneuron lamination in the cortex
Collapse
|
123
|
Abstract
Microglia and non-parenchymal macrophages in the brain are mononuclear phagocytes that are increasingly recognized to be essential players in the development, homeostasis and diseases of the central nervous system. With the availability of new genetic, molecular and pharmacological tools, considerable advances have been made towards our understanding of the embryonic origins, developmental programmes and functions of these cells. These exciting discoveries, some of which are still controversial, also raise many new questions, which makes brain macrophage biology a fast-growing field at the intersection of neuroscience and immunology. Here, we review the current knowledge of how and where brain macrophages are generated, with a focus on parenchymal microglia. We also discuss their normal functions during development and homeostasis, the disturbance of which may lead to various neurodegenerative and neuropsychiatric diseases.
Collapse
|
124
|
Thion MS, Garel S. On place and time: microglia in embryonic and perinatal brain development. Curr Opin Neurobiol 2017; 47:121-130. [PMID: 29080445 DOI: 10.1016/j.conb.2017.10.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/27/2017] [Accepted: 10/07/2017] [Indexed: 12/26/2022]
Abstract
Microglia, the brain-resident macrophages, play key roles in regulating synapse density and homeostasis in the postnatal and adult brain. However, microglia enter the brain during embryogenesis and recent studies have revealed additional early functions of these immune cells in prenatal and perinatal cerebral development. Such findings are of importance since prenatal inflammation and microglia dysfunction have been associated with several neurodevelopmental disorders. This review provides a selective overview of the early roles of microglia, their link with a specific spatiotemporal distribution and how they can be modulated by intrinsic factors or environmental signals.
Collapse
Affiliation(s)
- Morgane Sonia Thion
- Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France.
| | - Sonia Garel
- Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France.
| |
Collapse
|
125
|
Konishi H, Kobayashi M, Kunisawa T, Imai K, Sayo A, Malissen B, Crocker PR, Sato K, Kiyama H. Siglec-H is a microglia-specific marker that discriminates microglia from CNS-associated macrophages and CNS-infiltrating monocytes. Glia 2017; 65:1927-1943. [PMID: 28836308 DOI: 10.1002/glia.23204] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/25/2017] [Accepted: 07/25/2017] [Indexed: 01/06/2023]
Abstract
Several types of myeloid cell are resident in the CNS. In the steady state, microglia are present in the CNS parenchyma, whereas macrophages reside in boundary regions of the CNS, such as perivascular spaces, the meninges and choroid plexus. In addition, monocytes infiltrate into the CNS parenchyma from circulation upon blood-brain barrier breakdown after CNS injury and inflammation. Although several markers, such as CD11b and ionized calcium-binding adapter molecule 1 (Iba1), are frequently used as microglial markers, they are also expressed by other types of myeloid cell and microglia-specific markers were not defined until recently. Previous transcriptome analyses of isolated microglia identified a transmembrane lectin, sialic acid-binding immunoglobulin-like lectin H (Siglec-H), as a molecular signature for microglia; however, this was not confirmed by histological studies in the nervous system and the reliability of Siglec-H as a microglial marker remained unclear. Here, we demonstrate that Siglec-H is an authentic marker for microglia in mice by immunohistochemistry using a Siglec-H-specific antibody. Siglec-H was expressed by parenchymal microglia from developmental stages to adulthood, and the expression was maintained in activated microglia under injury or inflammatory condition. However, Siglec-H expression was absent from CNS-associated macrophages and CNS-infiltrating monocytes, except for a minor subset of cells. We also show that the Siglech gene locus is a feasible site for specific targeting of microglia in the nervous system. In conclusion, Siglec-H is a reliable marker for microglia that will allow histological identification of microglia and microglia-specific gene manipulation in the nervous system.
Collapse
Affiliation(s)
- Hiroyuki Konishi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Masaaki Kobayashi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Taikan Kunisawa
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Kenta Imai
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Akira Sayo
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.,Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS UMR, Marseille, 13288, France
| | - Paul R Crocker
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Katsuaki Sato
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, 889-1692, Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| |
Collapse
|
126
|
Martín-Estebané M, Navascués J, Sierra-Martín A, Martín-Guerrero SM, Cuadros MA, Carrasco MC, Marín-Teva JL. Onset of microglial entry into developing quail retina coincides with increased expression of active caspase-3 and is mediated by extracellular ATP and UDP. PLoS One 2017; 12:e0182450. [PMID: 28763502 PMCID: PMC5538646 DOI: 10.1371/journal.pone.0182450] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 07/18/2017] [Indexed: 12/31/2022] Open
Abstract
Microglial cell precursors located in the area of the base of the pecten and the optic nerve head (BP/ONH) start to enter the retina of quail embryos at the 7th day of incubation (E7), subsequently colonizing the entire retina by central-to-peripheral tangential migration, as previously shown by our group. The present study demonstrates a precise chronological coincidence of the onset of microglial cell entry into the retina with a striking increase in death of retinal cells, as revealed by their active caspase-3 expression and TUNEL staining, in regions dorsal to the BP/ONH area, suggesting that dying retinal cells would contribute to the microglial cell inflow into the retina. However, the molecular mechanisms involved in this inflow are currently unclear. Extracellular nucleotides, such as ATP and UDP, have previously been shown to favor migration of microglia towards brain injuries because they are released by apoptotic cells and stimulate both chemotaxis and chemokinesis in microglial cells via signaling through purinergic receptors. Hence, we tested here the hypothesis that ATP and UDP play a role in the entry and migration of microglial precursors into the developing retina. For this purpose, we used an experimental model system based on organotypic cultures of E6.5 quail embryo retina explants, which mimics the entry and migration of microglial precursors in the in situ developing retina. Inhibition of purinergic signaling by treating retina explants with either apyrase, a nucleotide-hydrolyzing enzyme, or suramin, a broad spectrum antagonist of purinergic receptors, significantly prevents the entry of microglial cells into the retina. In addition, treatment of retina explants with either exogenous ATP or UDP results in significantly increased numbers of microglial cells entering the retina. In light of these findings, we conclude that purinergic signaling by extracellular ATP and UDP is necessary for the entry and migration of microglial cells into the embryonic retina by inducing chemokinesis in these cells.
Collapse
Affiliation(s)
- María Martín-Estebané
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Julio Navascués
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Ana Sierra-Martín
- Departamento de Biología Celular, Fisiología e Inmunología, Facultad de Biociencias, Universidad Autónoma de Barcelona, Bellaterra, Barcelona, Spain
| | | | - Miguel A. Cuadros
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - María-Carmen Carrasco
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - José L. Marín-Teva
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
- * E-mail:
| |
Collapse
|
127
|
Wong EL, Stowell RD, Majewska AK. What the Spectrum of Microglial Functions Can Teach us About Fetal Alcohol Spectrum Disorder. Front Synaptic Neurosci 2017; 9:11. [PMID: 28674490 PMCID: PMC5474469 DOI: 10.3389/fnsyn.2017.00011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/29/2017] [Indexed: 12/18/2022] Open
Abstract
Alcohol exposure during gestation can lead to severe defects in brain development and lifelong physical, behavioral and learning deficits that are classified under the umbrella term fetal alcohol spectrum disorder (FASD). Sadly, FASD is diagnosed at an alarmingly high rate, affecting 2%-5% of live births in the United States, making it the most common non-heritable cause of mental disability. Currently, no standard therapies exist that are effective at battling FASD symptoms, highlighting a pressing need to better understand the underlying mechanisms by which alcohol affects the developing brain. While it is clear that sensory and cognitive deficits are driven by inappropriate development and remodeling of the neural circuits that mediate these processes, alcohol's actions acutely and long-term on the brain milieu are diverse and complex. Microglia, the brain's immune cells, have been thought to be a target for alcohol during development because of their exquisite ability to rapidly detect and respond to perturbations affecting the brain. Additionally, our view of these immune cells is rapidly changing, and recent studies have revealed a myriad of microglial physiological functions critical for normal brain development and long-term function. A clear and complete understanding of how microglial roles on this end of the spectrum may be altered in FASD is currently lacking. Such information could provide important insights toward novel therapeutic targets for FASD treatment. Here we review the literature that links microglia to neural circuit remodeling and provide a discussion of the current understanding of how developmental alcohol exposure affects microglial behavior in the context of developing brain circuits.
Collapse
Affiliation(s)
- Elissa L. Wong
- Department of Environmental Medicine, University of Rochester Medical CenterRochester, NY, United States
| | - Rianne D. Stowell
- Department of Neuroscience, University of Rochester Medical CenterRochester, NY, United States
| | - Ania K. Majewska
- Department of Neuroscience, University of Rochester Medical CenterRochester, NY, United States
| |
Collapse
|
128
|
Abstract
Microglia begin colonizing the developing brain as early as embryonic day 9, prior to the emergence of neurons and other glia. Their ontogeny is also distinct from other central nervous system cells, as they derive from yolk sac hematopoietic progenitors and not neural progenitors. In this review, we feature these unique characteristics of microglia and assess the spatiotemporal similarities between microglia colonization of the central nervous system and embryonic neurogenesis. We also infer to existing evidence for microglia function from embryonic through to postnatal neurodevelopment to postulate roles for microglia in neurogenesis.
Collapse
Affiliation(s)
- Chih Kong Tong
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Malaysia
| | - Sharmili Vidyadaran
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Malaysia
| |
Collapse
|
129
|
Walsh CE, Hitchcock PF. Progranulin regulates neurogenesis in the developing vertebrate retina. Dev Neurobiol 2017; 77:1114-1129. [PMID: 28380680 PMCID: PMC5568971 DOI: 10.1002/dneu.22499] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022]
Abstract
We evaluated the expression and function of the microglia‐specific growth factor, Progranulin‐a (Pgrn‐a) during developmental neurogenesis in the embryonic retina of zebrafish. At 24 hpf pgrn‐a is expressed throughout the forebrain, but by 48 hpf pgrn‐a is exclusively expressed by microglia and/or microglial precursors within the brain and retina. Knockdown of Pgrn‐a does not alter the onset of neurogenic programs or increase cell death, however, in its absence, neurogenesis is significantly delayed—retinal progenitors fail to exit the cell cycle at the appropriate developmental time and postmitotic cells do not acquire markers of terminal differentiation, and microglial precursors do not colonize the retina. Given the link between Progranulin and cell cycle regulation in peripheral tissues and transformed cells, we analyzed cell cycle kinetics among retinal progenitors following Pgrn‐a knockdown. Depleting Pgrn‐a results in a significant lengthening of the cell cycle. These data suggest that Pgrn‐a plays a dual role during nervous system development by governing the rate at which progenitors progress through the cell cycle and attracting microglial progenitors into the embryonic brain and retina. Collectively, these data show that Pgrn‐a governs neurogenesis by regulating cell cycle kinetics and the transition from proliferation to cell cycle exit and differentiation. © 2017 The Authors. Developmental Neurobiology Published by Wiley Periodicals, Inc. Develop Neurobiol 77: 1114–1129, 2017
Collapse
Affiliation(s)
- Caroline E Walsh
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan, 48105.,Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, 48105
| | - Peter F Hitchcock
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan, 48105.,Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, 48105
| |
Collapse
|
130
|
Smolders SMT, Swinnen N, Kessels S, Arnauts K, Smolders S, Le Bras B, Rigo JM, Legendre P, Brône B. Age-specific function of α5β1 integrin in microglial migration during early colonization of the developing mouse cortex. Glia 2017; 65:1072-1088. [PMID: 28417486 DOI: 10.1002/glia.23145] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/10/2017] [Accepted: 03/14/2017] [Indexed: 12/31/2022]
Abstract
Microglia, the immune cells of the central nervous system, take part in brain development and homeostasis. They derive from primitive myeloid progenitors that originate in the yolk sac and colonize the brain mainly through intensive migration. During development, microglial migration speed declines which suggests that their interaction with the microenvironment changes. However, the matrix-cell interactions allowing dispersion within the parenchyma are unknown. Therefore, we aimed to better characterize the migration behavior and to assess the role of matrix-integrin interactions during microglial migration in the embryonic brain ex vivo. We focused on microglia-fibronectin interactions mediated through the fibronectin receptor α5β1 integrin because in vitro work indirectly suggested a role for this ligand-receptor pair. Using 2-photon time-lapse microscopy on acute ex vivo embryonic brain slices, we found that migration occurs in a saltatory pattern and is developmentally regulated. Most importantly, there is an age-specific function of the α5β1 integrin during microglial cortex colonization. At embryonic day (E) 13.5, α5β1 facilitates migration while from E15.5, it inhibits migration. These results indicate a developmentally regulated function of α5β1 integrin in microglial migration during colonization of the embryonic brain.
Collapse
Affiliation(s)
- Sophie Marie-Thérèse Smolders
- UHasselt, BIOMED, Diepenbeek, Belgium.,INSERM, UMR_S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | | | | | | - Silke Smolders
- UHasselt, BIOMED, Diepenbeek, Belgium.,Laboratory of Neuronal Differentiation, VIB Center for the Biology of Disease, Leuven and Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Barbara Le Bras
- INSERM, UMR_S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | | - Pascal Legendre
- INSERM, UMR_S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | |
Collapse
|
131
|
Mosser CA, Baptista S, Arnoux I, Audinat E. Microglia in CNS development: Shaping the brain for the future. Prog Neurobiol 2017; 149-150:1-20. [DOI: 10.1016/j.pneurobio.2017.01.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 12/22/2022]
|
132
|
Ibañez Rodriguez MP, Noctor SC, Muñoz EM. Cellular Basis of Pineal Gland Development: Emerging Role of Microglia as Phenotype Regulator. PLoS One 2016; 11:e0167063. [PMID: 27861587 PMCID: PMC5115862 DOI: 10.1371/journal.pone.0167063] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/08/2016] [Indexed: 12/04/2022] Open
Abstract
The adult pineal gland is composed of pinealocytes, astrocytes, microglia, and other interstitial cells that have been described in detail. However, factors that contribute to pineal development have not been fully elucidated, nor have pineal cell lineages been well characterized. We applied systematic double, triple and quadruple labeling of cell-specific markers on prenatal, postnatal and mature rat pineal gland tissue combined with confocal microscopy to provide a comprehensive view of the cellular dynamics and cell lineages that contribute to pineal gland development. The pineal gland begins as an evagination of neuroepithelium in the roof of the third ventricle. The pineal primordium initially consists of radially aligned Pax6+ precursor cells that express vimentin and divide at the ventricular lumen. After the tubular neuroepithelium fuses, the distribution of Pax6+ cells transitions to include rosette-like structures and later, dispersed cells. In the developing gland all dividing cells express Pax6, indicating that Pax6+ precursor cells generate pinealocytes and some interstitial cells. The density of Pax6+ cells decreases across pineal development as a result of cellular differentiation and microglial phagocytosis, but Pax6+ cells remain in the adult gland as a distinct population. Microglial colonization begins after pineal recess formation. Microglial phagocytosis of Pax6+ cells is not common at early stages but increases as microglia colonize the gland. In the postnatal gland microglia affiliate with Tuj1+ nerve fibers, IB4+ blood vessels, and Pax6+ cells. We demonstrate that microglia engulf Pax6+ cells, nerve fibers, and blood vessel-related elements, but not pinealocytes. We conclude that microglia play a role in pineal gland formation and homeostasis by regulating the precursor cell population, remodeling blood vessels and pruning sympathetic nerve fibers.
Collapse
Affiliation(s)
- María P. Ibañez Rodriguez
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo, National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
| | - Stephen C. Noctor
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California, Davis, School of Medicine, Sacramento, CA, United States of America
- * E-mail: (EMM); (SCN)
| | - Estela M. Muñoz
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo, National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
- * E-mail: (EMM); (SCN)
| |
Collapse
|
133
|
Reemst K, Noctor SC, Lucassen PJ, Hol EM. The Indispensable Roles of Microglia and Astrocytes during Brain Development. Front Hum Neurosci 2016; 10:566. [PMID: 27877121 PMCID: PMC5099170 DOI: 10.3389/fnhum.2016.00566] [Citation(s) in RCA: 357] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 10/25/2016] [Indexed: 01/17/2023] Open
Abstract
Glia are essential for brain functioning during development and in the adult brain. Here, we discuss the various roles of both microglia and astrocytes, and their interactions during brain development. Although both cells are fundamentally different in origin and function, they often affect the same developmental processes such as neuro-/gliogenesis, angiogenesis, axonal outgrowth, synaptogenesis and synaptic pruning. Due to their important instructive roles in these processes, dysfunction of microglia or astrocytes during brain development could contribute to neurodevelopmental disorders and potentially even late-onset neuropathology. A better understanding of the origin, differentiation process and developmental functions of microglia and astrocytes will help to fully appreciate their role both in the developing as well as in the adult brain, in health and disease.
Collapse
Affiliation(s)
- Kitty Reemst
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| | - Stephen C. Noctor
- Department of Psychiatry and Behavioral Sciences, UC Davis MIND InstituteSacramento, CA, USA
| | - Paul J. Lucassen
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| | - Elly M. Hol
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
- Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands
- Netherlands Institute for NeuroscienceAmsterdam, Netherlands
| |
Collapse
|
134
|
Multitasking Microglia and Alzheimer's Disease: Diversity, Tools and Therapeutic Targets. J Mol Neurosci 2016; 60:390-404. [PMID: 27660215 DOI: 10.1007/s12031-016-0825-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 08/17/2016] [Indexed: 01/08/2023]
Abstract
Given the importance of microglia to inflammatory, phagocytic and synaptic modulatory processes, their function is vital in physiological and pathological brain. The impairment of microglia in Alzheimer's disease has been demonstrated on genetic, epigenetic, transcriptional and functional levels using unbiased systems level approaches. Recent studies have highlighted the immense phenotypic diversity of microglia, including the ability to adopt distinct and dynamic phenotypes in ageing and disease. We review the origins and functions of healthy microglia and the established and emerging models and techniques available for their study. Furthermore, we highlight recent advances on the role, heterogeneity and dysfunction of microglia in Alzheimer's disease and discuss the potential for therapeutic interventions targeting microglia. Microglia-selective molecular fingerprints will guide detailed functional analysis of microglial subsets and may aid in the development of therapies specifically targeting microglia.
Collapse
|
135
|
Infiltrating cells from host brain restore the microglial population in grafted cortical tissue. Sci Rep 2016; 6:33080. [PMID: 27615195 PMCID: PMC5018877 DOI: 10.1038/srep33080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/19/2016] [Indexed: 12/20/2022] Open
Abstract
Transplantation of embryonic cortical tissue is considered as a promising therapy for brain injury. Grafted neurons can reestablish neuronal network and improve cortical function of the host brain. Microglia is a key player in regulating neuronal survival and plasticity, but its activation and dynamics in grafted cortical tissue remain unknown. Using two-photon intravital imaging and parabiotic model, here we investigated the proliferation and source of microglia in the donor region by transplanting embryonic cortical tissue into adult cortex. Live imaging showed that the endogenous microglia of the grafted tissue were rapidly lost after transplantation. Instead, host-derived microglia infiltrated and colonized the graft. Parabiotic model suggested that the main source of infiltrating cells is the parenchyma of the host brain. Colonized microglia proliferated and experienced an extensive morphological transition and eventually differentiated into resting ramified morphology. Collectively, these results demonstrated that donor tissue has little contribution to the activated microglia and host brain controls the microglial population in the graft.
Collapse
|
136
|
Eyo UB, Miner SA, Weiner JA, Dailey ME. Developmental changes in microglial mobilization are independent of apoptosis in the neonatal mouse hippocampus. Brain Behav Immun 2016; 55:49-59. [PMID: 26576723 PMCID: PMC4864211 DOI: 10.1016/j.bbi.2015.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/09/2015] [Accepted: 11/09/2015] [Indexed: 12/31/2022] Open
Abstract
During CNS development, microglia transform from highly mobile amoeboid-like cells to primitive ramified forms and, finally, to highly branched but relatively stationary cells in maturity. The factors that control developmental changes in microglia are largely unknown. Because microglia detect and clear apoptotic cells, developmental changes in microglia may be controlled by neuronal apoptosis. Here, we assessed the extent to which microglial cell density, morphology, motility, and migration are regulated by developmental apoptosis, focusing on the first postnatal week in the mouse hippocampus when the density of apoptotic bodies peaks at postnatal day 4 and declines sharply thereafter. Analysis of microglial form and distribution in situ over the first postnatal week showed that, although there was little change in the number of primary microglial branches, microglial cell density increased significantly, and microglia were often seen near or engulfing apoptotic bodies. Time-lapse imaging in hippocampal slices harvested at different times over the first postnatal week showed differences in microglial motility and migration that correlated with the density of apoptotic bodies. The extent to which these changes in microglia are driven by developmental neuronal apoptosis was assessed in tissues from BAX null mice lacking apoptosis. We found that apoptosis can lead to local microglial accumulation near apoptotic neurons in the pyramidal cell body layer but, unexpectedly, loss of apoptosis did not alter overall microglial cell density in vivo or microglial motility and migration in ex vivo tissue slices. These results demonstrate that developmental changes in microglial form, distribution, motility, and migration occur essentially normally in the absence of developmental apoptosis, indicating that factors other than neuronal apoptosis regulate these features of microglial development.
Collapse
|
137
|
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: 390] [Impact Index Per Article: 43.3] [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
|
138
|
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
|
139
|
Smolders S, Smolders SMT, Swinnen N, Gärtner A, Rigo JM, Legendre P, Brône B. Maternal immune activation evoked by polyinosinic:polycytidylic acid does not evoke microglial cell activation in the embryo. Front Cell Neurosci 2015; 9:301. [PMID: 26300736 PMCID: PMC4525016 DOI: 10.3389/fncel.2015.00301] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/22/2015] [Indexed: 12/13/2022] Open
Abstract
Several studies have indicated that inflammation during pregnancy increases the risk for the development of neuropsychiatric disorders in the offspring. Morphological brain abnormalities combined with deviations in the inflammatory status of the brain can be observed in patients of both autism and schizophrenia. It was shown that acute infection can induce changes in maternal cytokine levels which in turn are suggested to affect fetal brain development and increase the risk on the development of neuropsychiatric disorders in the offspring. Animal models of maternal immune activation reproduce the etiology of neurodevelopmental disorders such as schizophrenia and autism. In this study the poly (I:C) model was used to mimic viral immune activation in pregnant mice in order to assess the activation status of fetal microglia in these developmental disorders. Because microglia are the resident immune cells of the brain they were expected to be activated due to the inflammatory stimulus. Microglial cell density and activation level in the fetal cortex and hippocampus were determined. Despite the presence of a systemic inflammation in the pregnant mice, there was no significant difference in fetal microglial cell density or immunohistochemically determined activation level between the control and inflammation group. These data indicate that activation of the fetal microglial cells is not likely to be responsible for the inflammation induced deficits in the offspring in this model.
Collapse
Affiliation(s)
- Silke Smolders
- BIOMED - Hasselt University Hasselt, Belgium ; Laboratory of Neuronal Differentiation, VIB Center for the Biology of Disease, Leuven and Center for Human Genetics, KU Leuven Leuven, Belgium
| | - Sophie M T Smolders
- BIOMED - Hasselt University Hasselt, Belgium ; INSERM, UMR S 1130, Université Pierre et Marie Curie Paris, France ; CNRS, UMR 8246, Université Pierre et Marie Curie Paris, France ; UM 119 NPS, Université Pierre et Marie Curie Paris, France
| | | | - Annette Gärtner
- Laboratory of Neuronal Differentiation, VIB Center for the Biology of Disease, Leuven and Center for Human Genetics, KU Leuven Leuven, Belgium
| | | | - Pascal Legendre
- INSERM, UMR S 1130, Université Pierre et Marie Curie Paris, France ; CNRS, UMR 8246, Université Pierre et Marie Curie Paris, France ; UM 119 NPS, Université Pierre et Marie Curie Paris, France
| | - Bert Brône
- BIOMED - Hasselt University Hasselt, Belgium
| |
Collapse
|
140
|
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
|
141
|
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
|
142
|
Ginhoux F, Prinz M. Origin of microglia: current concepts and past controversies. Cold Spring Harb Perspect Biol 2015; 7:a020537. [PMID: 26134003 DOI: 10.1101/cshperspect.a020537] [Citation(s) in RCA: 286] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS), which sit in close proximity to neural structures and are intimately involved in brain homeostasis. The microglial population also plays fundamental roles during neuronal expansion and differentiation, as well as in the perinatal establishment of synaptic circuits. Any change in the normal brain environment results in microglial activation, which can be detrimental if not appropriately regulated. Aberrant microglial function has been linked to the development of several neurological and psychiatric diseases. However, microglia also possess potent immunoregulatory and regenerative capacities, making them attractive targets for therapeutic manipulation. Such rationale manipulations will, however, require in-depth knowledge of their origins and the molecular mechanisms underlying their homeostasis. Here, we discuss the latest advances in our understanding of the origin, differentiation, and homeostasis of microglial cells and their myelomonocytic relatives in the CNS.
Collapse
Affiliation(s)
- Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, 79106 Freiburg, Germany BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
143
|
Harris J, Tomassy GS, Arlotta P. Building blocks of the cerebral cortex: from development to the dish. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:529-44. [PMID: 25926310 DOI: 10.1002/wdev.192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/23/2015] [Accepted: 03/23/2015] [Indexed: 12/19/2022]
Abstract
Since Ramon y Cajal's examination of the cellular makeup of the cerebral cortex, it has been appreciated that this tissue exhibits some of the greatest degrees of cellular heterogeneity in the entire nervous system. This intricate structure emerges during a well-choreographed developmental process. Here, we review current classifications of the cellular constituents of the cerebral cortex and examine how these building blocks are forged during development. We also look at how basic developmental features underlying cortex formation in vivo have been applied to protocols aimed at generating cortical tissue in vitro.
Collapse
Affiliation(s)
- James Harris
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Giulio Srubek Tomassy
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| |
Collapse
|
144
|
Bjornsson CS, Apostolopoulou M, Tian Y, Temple S. It takes a village: constructing the neurogenic niche. Dev Cell 2015; 32:435-46. [PMID: 25710530 DOI: 10.1016/j.devcel.2015.01.010] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Although many features of neurogenesis during development and in the adult are intrinsic to the neurogenic cells themselves, the role of the microenvironment is irrefutable. The neurogenic niche is a melting pot of cells and factors that influence CNS development. How do the diverse elements assemble and when? How does the niche change structurally and functionally during embryogenesis and in adulthood? In this review, we focus on the impact of non-neural cells that participate in the neurogenic niche, highlighting how cells of different embryonic origins influence this critical germinal space.
Collapse
Affiliation(s)
| | | | - Yangzi Tian
- SUNY Polytechnic Institute, College of Nanoscale Science and Engineering, Albany, NY 12203, USA
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| |
Collapse
|
145
|
Neural progenitor cells orchestrate microglia migration and positioning into the developing cortex. Nat Commun 2014; 5:5611. [PMID: 25425146 DOI: 10.1038/ncomms6611] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 10/20/2014] [Indexed: 12/26/2022] Open
Abstract
Microglia are observed in the early developing forebrain and contribute to the regulation of neurogenesis through still unravelled mechanisms. In the developing cerebral cortex, microglia cluster in the ventricular/subventricular zone (VZ/SVZ), a region containing Cxcl12-expressing basal progenitors (BPs). Here we show that the ablation of BP as well as genetic loss of Cxcl12 affect microglia recruitment into the SVZ. Ectopic Cxcl12 expression or pharmacological blockage of CxcR4 further supports that Cxcl12/CxcR4 signalling is involved in microglial recruitment during cortical development. Furthermore, we found that cell death in the developing forebrain triggers microglial proliferation and that this is mediated by the release of macrophage migration inhibitory factor (MIF). Finally, we show that the depletion of microglia in mice lacking receptor for colony-stimulating factor-1 (Csf-1R) reduces BPs into the cerebral cortex.
Collapse
|
146
|
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: 416] [Impact Index Per Article: 37.8] [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
|
147
|
Sierra A, Navascués J, Cuadros MA, Calvente R, Martín-Oliva D, Ferrer-Martín RM, Martín-Estebané M, Carrasco MC, Marín-Teva JL. Expression of inducible nitric oxide synthase (iNOS) in microglia of the developing quail retina. PLoS One 2014; 9:e106048. [PMID: 25170849 PMCID: PMC4149512 DOI: 10.1371/journal.pone.0106048] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/28/2014] [Indexed: 12/17/2022] Open
Abstract
Inducible nitric oxide synthase (iNOS), which produce large amounts of nitric oxide (NO), is induced in macrophages and microglia in response to inflammatory mediators such as LPS and cytokines. Although iNOS is mainly expressed by microglia that become activated in different pathological and experimental situations, it was recently reported that undifferentiated amoeboid microglia can also express iNOS during normal development. The aim of this study was to investigate the pattern of iNOS expression in microglial cells during normal development and after their activation with LPS by using the quail retina as model. iNOS expression was analyzed by iNOS immunolabeling, western-blot, and RT-PCR. NO production was determined by using DAR-4M AM, a reliable fluorescent indicator of subcellular NO production by iNOS. Embryonic, postnatal, and adult in situ quail retinas were used to analyze the pattern of iNOS expression in microglial cells during normal development. iNOS expression and NO production in LPS-treated microglial cells were investigated by an in vitro approach based on organotypic cultures of E8 retinas, in which microglial cell behavior is similar to that of the in situ retina, as previously demonstrated in our laboratory. We show here that amoeboid microglia in the quail retina express iNOS during normal development. This expression is stronger in microglial cells migrating tangentially in the vitreal part of the retina and is downregulated, albeit maintained, when microglia differentiate and become ramified. LPS treatment of retina explants also induces changes in the morphology of amoeboid microglia compatible with their activation, increasing their lysosomal compartment and upregulating iNOS expression with a concomitant production of NO. Taken together, our findings demonstrate that immature microglial cells express iNOS during normal development, suggesting a certain degree of activation. Furthermore, LPS treatment induces overactivation of amoeboid microglia, resulting in a significant iNOS upregulation.
Collapse
Affiliation(s)
- Ana Sierra
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Julio Navascués
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Miguel A. Cuadros
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Ruth Calvente
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - David Martín-Oliva
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - Rosa M. Ferrer-Martín
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - María Martín-Estebané
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - María-Carmen Carrasco
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
| | - José L. Marín-Teva
- Departamento de Biología Celular, Facultad de Ciencias, Universidad de Granada, Granada, Spain
- * E-mail:
| |
Collapse
|
148
|
Haw RTY, Tong CK, Yew A, Lee HC, Phillips JB, Vidyadaran S. A three-dimensional collagen construct to model lipopolysaccharide-induced activation of BV2 microglia. J Neuroinflammation 2014; 11:134. [PMID: 25074682 PMCID: PMC4128540 DOI: 10.1186/1742-2094-11-134] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 07/16/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND We report a novel method of culturing microglia in three dimension (3D) using collagen as a substrate. By culturing microglia within a matrix, we aim to emulate the physical state of microglia embedded within parenchyma. METHODS BV2 microglia cell suspensions were prepared with type I collagen and cast into culture plates. To characterise the BV2 microglia cultured in 3D, the cultures were evaluated for their viability, cell morphology and response to lipopolysaccharide (LPS) activation. Conventional monolayer cultures (grown on uncoated and collagen-coated polystyrene) were set up concurrently for comparison. RESULTS BV2 microglia in 3D collagen matrices were viable at 48 hrs of culture and exhibit a ramified morphology with multiplanar cytoplasmic projections. Following stimulation with 1 μg/ml LPS, microglia cultured in 3D collagen gels increase their expression of nitric oxide (NO) and CD40, indicating their capacity to become activated within the matrix. Up to 97.8% of BV2 microglia grown in 3D cultures gained CD40 positivity in response to LPS, compared to approximately 60% of cells grown in a monolayer (P<.05). BV2 microglia in 3D collagen gels also showed increased mRNA and protein expression of inflammatory cytokines IL-6, TNF-α and the chemoattractant MCP-1 following LPS stimulation. CONCLUSIONS In summary, BV2 microglia cultured in 3D collagen hydrogels exhibit multiplanar cytoplasmic projections and undergo a characteristic and robust activation response to LPS. This culture system is accessible to a wide range of analyses and provides a useful new in vitro tool for research into microglial activation.
Collapse
Affiliation(s)
- Randy Tatt Yhew Haw
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Chih Kong Tong
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Andrew Yew
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Han Chung Lee
- Genetic & Regenerative Medicine Research Centre (GRMRC) & Department of Obstetrics & Gynaecology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - James B Phillips
- Department of Biomaterials & Tissue Engineering, University College London, UCL Eastman Dental Institute, 256 Gray’s Inn Road, London WC1X 8LD, UK
| | - Sharmili Vidyadaran
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
149
|
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: 63] [Impact Index Per Article: 5.7] [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
|
150
|
Legendre P, Le Corronc H. [Microglial cells and development of the embryonic central nervous system]. Med Sci (Paris) 2014; 30:147-52. [PMID: 24572112 DOI: 10.1051/medsci/20143002011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Microglia cells are the macrophages of the central nervous system with a crucial function in the homeostasis of the adult brain. However, recent studies showed that microglial cells may also have important functions during early embryonic central nervous system development. In this review we summarize recent works on the extra embryonic origin of microglia, their progenitor niche, the pattern of their invasion of the embryonic central nervous system and on interactions between embryonic microglia and their local environment during invasion. We describe microglial functions during development of embryonic neuronal networks, including their roles in neurogenesis, in angiogenesis and developmental cell death. These recent discoveries open a new field of research on the functions of neural-microglial interactions during the development of the embryonic central nervous system.
Collapse
Affiliation(s)
- Pascal Legendre
- Inserm UMR-S 1130, université Pierre et Marie Curie (UPMC), 9, quai Saint-Bernard, Paris, France - CNRS, UMR 8246, UPMC, 9, quai Saint-Bernard, Paris, France - UPMC UM CR18, 9, quai Saint-Bernard, Paris, France
| | - Hervé Le Corronc
- Inserm UMR-S 1130, université Pierre et Marie Curie (UPMC), 9, quai Saint-Bernard, Paris, France - CNRS, UMR 8246, UPMC, 9, quai Saint-Bernard, Paris, France - UPMC UM CR18, 9, quai Saint-Bernard, Paris, France - Université d'Angers, 40, rue de Rennes, Angers, France
| |
Collapse
|