751
|
Copland DA, Theodoropoulou S, Liu J, Dick AD. A Perspective of AMD Through the Eyes of Immunology. ACTA ACUST UNITED AC 2018; 59:AMD83-AMD92. [DOI: 10.1167/iovs.18-23893] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- David A. Copland
- Translational Health Sciences (Ophthalmology), University of Bristol, Bristol, United Kingdom
- National Institute for Health Research Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital and University College London-Institute of Ophthalmology, London, United Kingdom
| | - Sofia Theodoropoulou
- Translational Health Sciences (Ophthalmology), University of Bristol, Bristol, United Kingdom
- Bristol Eye Hospital, Bristol, United Kingdom
| | - Jian Liu
- Translational Health Sciences (Ophthalmology), University of Bristol, Bristol, United Kingdom
| | - Andrew D. Dick
- Translational Health Sciences (Ophthalmology), University of Bristol, Bristol, United Kingdom
- National Institute for Health Research Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital and University College London-Institute of Ophthalmology, London, United Kingdom
- Bristol Eye Hospital, Bristol, United Kingdom
- University College London–Institute of Ophthalmology, London, United Kingdom
| |
Collapse
|
752
|
Abstract
Glial cell types were classified less than 100 years ago by del Rio-Hortega. For instance, he correctly surmised that microglia in pathologic central nervous system (CNS) were "voracious monsters" that helped clean the tissue. Although these historical predictions were remarkably accurate, innovative technologies have revealed novel molecular, cellular, and dynamic physiologic aspects of CNS glia. In this review, we integrate recent findings regarding the roles of glia and glial interactions in healthy and injured spinal cord. The three major glial cell types are considered in healthy CNS and after spinal cord injury (SCI). Astrocytes, which in the healthy CNS regulate neurotransmitter and neurovascular dynamics, respond to SCI by becoming reactive and forming a glial scar that limits pathology and plasticity. Microglia, which in the healthy CNS scan for infection/damage, respond to SCI by promoting axon growth and remyelination-but also with hyperactivation and cytotoxic effects. Oligodendrocytes and their precursors, which in healthy tissue speed axon conduction and support axonal function, respond to SCI by differentiating and producing myelin, but are susceptible to death. Thus, post-SCI responses of each glial cell can simultaneously stimulate and stifle repair. Interestingly, potential therapies could also target interactions between these cells. Astrocyte-microglia cross-talk creates a feed-forward loop, so shifting the response of either cell could amplify repair. Astrocytes, microglia, and oligodendrocytes/precursors also influence post-SCI cell survival, differentiation, and remyelination, as well as axon sparing. Therefore, optimizing post-SCI responses of glial cells-and interactions between these CNS cells-could benefit neuroprotection, axon plasticity, and functional recovery.
Collapse
Affiliation(s)
- Andrew D Gaudet
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO, 80309, USA.
- Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO, 80309, USA.
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
753
|
Tenner AJ, Stevens B, Woodruff TM. New tricks for an ancient system: Physiological and pathological roles of complement in the CNS. Mol Immunol 2018; 102:3-13. [PMID: 29958698 DOI: 10.1016/j.molimm.2018.06.264] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022]
Abstract
While the mechanisms underlying the functions of the complement system in the central nervous system (CNS) and systemically, namely opsonization, chemotaxis, membrane lysis, and regulation of inflammation are the same, the plethora of functions that complement orchestrates in the central nervous system (CNS) is complex. Strictly controlled expression of complement effector molecules, regulators and receptors across the gamut of life stages (embryogenesis, development and maturation, aging and disease) dictate fascinating contributions for this ancient system. Furthermore, it is becoming apparent that complement functions differ widely across distinct brain regions. This review provides a comprehensive overview of the newly identified roles for complement in the brain, including its roles in CNS development and function, during aging and in the processes of neurodegeneration. The diversity and selectively of beneficial and detrimental activities of complement, while challenging, should lead to precision targeting of specific components to provide disease modifying treatments for devastating psychiatric and neurodegenerative disorders that are still without effective treatment.
Collapse
Affiliation(s)
- Andrea J Tenner
- Departments of Molecular Biology and Biochemistry, Neurobiology and Behavior, and Pathology and Laboratory Medicine, University of California, Irvine, CA, United States.
| | - Beth Stevens
- F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Department of Neurobiology, Harvard Medical School, Boston, MA, United States; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
754
|
Rosin JM, Kurrasch DM. In utero electroporation induces cell death and alters embryonic microglia morphology and expression signatures in the developing hypothalamus. J Neuroinflammation 2018; 15:181. [PMID: 29895301 PMCID: PMC5998590 DOI: 10.1186/s12974-018-1213-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 05/21/2018] [Indexed: 12/25/2022] Open
Abstract
Background Since its inception in 2001, in utero electroporation (IUE) has been widely used by the neuroscience community. IUE is a technique developed to introduce plasmid DNA into embryonic mouse brains without permanently removing the embryos from the uterus. Given that IUE labels cells that line the ventricles, including radial fibers and migrating neuroblasts, this technique is an excellent tool for studying factors that govern neural cell fate determination and migration in the developing mouse brain. Whether IUE has an effect on microglia, the immune cells of the central nervous system (CNS), has yet to be investigated. Methods We used IUE and the pCIG2, pCIC-Ascl1, or pRFP-C-RS expression vectors to label radial glia lining the ventricles of the embryonic cortex and/or hypothalamus. Specifically, we conducted IUE at E14.5 and harvested the brains at E15.5 or E17.5. Immunohistochemistry, along with cytokine and chemokine analyses, were performed on embryonic brains with or without IUE exposure. Results IUE using the pCIG2, pCIC-Ascl1, or pRFP-C-RS vectors alone altered microglia morphology, where the majority of microglia near the ventricles were amoeboid and displayed altered expression signatures, including the upregulation of Cd45 and downregulation of P2ry12. Moreover, IUE led to increases in P2ry12− cells that were Iba1+/IgG+ double-positive in the brain parenchyma and resembled macrophages infiltrating the brain proper from the periphery. Furthermore, IUE resulted in a significant increase in cell death in the developing hypothalamus, with concomitant increases in cytokines and chemokines known to be released during pro-inflammatory states (IL-1β, IL-6, MIP-2, RANTES, MCP-1). Interestingly, the cortex was protected from elevated cell death following IUE, implying that microglia that reside in the hypothalamus might be particularly sensitive during embryonic development. Conclusions Our results suggest that IUE might have unintended consequences of activating microglia in the embryonic brain, which could have long-term effects, particularly within the hypothalamus. Electronic supplementary material The online version of this article (10.1186/s12974-018-1213-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jessica M Rosin
- Department of Medical Genetics, Cummings School of Medicine, University of Calgary, 3330 Hospital Drive NW, Room HS2215, Calgary, Alberta, T2N 4N1, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cummings School of Medicine, University of Calgary, 3330 Hospital Drive NW, Room HS2215, Calgary, Alberta, T2N 4N1, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada. .,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
755
|
Tasic B. Single cell transcriptomics in neuroscience: cell classification and beyond. Curr Opin Neurobiol 2018; 50:242-249. [DOI: 10.1016/j.conb.2018.04.021] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/15/2022]
|
756
|
Lopez‐Atalaya JP, Askew KE, Sierra A, Gomez‐Nicola D. Development and maintenance of the brain's immune toolkit: Microglia and non-parenchymal brain macrophages. Dev Neurobiol 2018; 78:561-579. [PMID: 29030904 PMCID: PMC6001428 DOI: 10.1002/dneu.22545] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/29/2017] [Accepted: 10/06/2017] [Indexed: 01/10/2023]
Abstract
Microglia and non-parenchymal macrophages located in the perivascular space, the meninges and the choroid plexus are independent immune populations that play vital roles in brain development, homeostasis, and tissue healing. Resident macrophages account for a significant proportion of cells in the brain and their density remains stable throughout the lifespan thanks to constant turnover. Microglia develop from yolk sac progenitors, later evolving through intermediate progenitors in a fine-tuned process in which intrinsic factors and external stimuli combine to progressively sculpt their cell type-specific transcriptional profiles. Recent evidence demonstrates that non-parenchymal macrophages are also generated during early embryonic development. In recent years, the development of powerful fate mapping approaches combined with novel genomic and transcriptomic methodologies have greatly expanded our understanding of how brain macrophages develop and acquire specialized functions, and how cell population dynamics are regulated. Here, we review the transcription factors, epigenetic remodeling, and signaling pathways orchestrating the embryonic development of microglia and non-parenchymal macrophages. Next, we describe the dynamics of the macrophage populations of the brain and discuss the role of progenitor cells, to gain a better understanding of their functions in the healthy and diseased brain. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 561-579, 2018.
Collapse
Affiliation(s)
- Jose P. Lopez‐Atalaya
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández‐Consejo Superior de Investigaciones Científicas (UMH‐CSIC), Avenida Ramón y Cajal, s/n, Sant Joan d'AlacantSpain
| | - Katharine E. Askew
- Southampton General Hospital, Biological Sciences, University of Southampton, South Lab&Path Block, LD80C, MP840SO166YDSouthamptonUnited Kingdom
| | - Amanda Sierra
- Achucarro Basque Center for NeuroscienceLeioa48940Spain
- Ikerbasque FoundationBilbao48013Spain
- University of the Basque Country EHU/UPVLeioa48940Spain
| | - Diego Gomez‐Nicola
- Southampton General Hospital, Biological Sciences, University of Southampton, South Lab&Path Block, LD80C, MP840SO166YDSouthamptonUnited Kingdom
| |
Collapse
|
757
|
Bennett FC, Bennett ML, Yaqoob F, Mulinyawe SB, Grant GA, Hayden Gephart M, Plowey ED, Barres BA. A Combination of Ontogeny and CNS Environment Establishes Microglial Identity. Neuron 2018; 98:1170-1183.e8. [PMID: 29861285 DOI: 10.1016/j.neuron.2018.05.014] [Citation(s) in RCA: 380] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/02/2018] [Accepted: 05/07/2018] [Indexed: 10/14/2022]
Abstract
Microglia, the brain's resident macrophages, are dynamic CNS custodians with surprising origins in the extra-embryonic yolk sac. The consequences of their distinct ontogeny are unknown but critical to understanding and treating brain diseases. We created a brain macrophage transplantation system to disentangle how environment and ontogeny specify microglial identity. We find that donor cells extensively engraft in the CNS of microglia-deficient mice, and even after exposure to a cell culture environment, microglia fully regain their identity when returned to the CNS. Though transplanted macrophages from multiple tissues can express microglial genes in the brain, only those of yolk-sac origin fully attain microglial identity. Transplanted macrophages of inappropriate origin, including primary human cells in a humanized host, express disease-associated genes and specific ontogeny markers. Through brain macrophage transplantation, we discover new principles of microglial identity that have broad applications to the study of disease and development of myeloid cell therapies.
Collapse
Affiliation(s)
- F Chris Bennett
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Mariko L Bennett
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Fazeela Yaqoob
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sara B Mulinyawe
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gerald A Grant
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Melanie Hayden Gephart
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Edward D Plowey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
758
|
Giera S, Luo R, Ying Y, Ackerman SD, Jeong SJ, Stoveken HM, Folts CJ, Welsh CA, Tall GG, Stevens B, Monk KR, Piao X. Microglial transglutaminase-2 drives myelination and myelin repair via GPR56/ADGRG1 in oligodendrocyte precursor cells. eLife 2018; 7:33385. [PMID: 29809138 PMCID: PMC5980231 DOI: 10.7554/elife.33385] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 05/18/2018] [Indexed: 12/29/2022] Open
Abstract
In the central nervous system (CNS), myelin formation and repair are regulated by oligodendrocyte (OL) lineage cells, which sense and integrate signals from their environment, including from other glial cells and the extracellular matrix (ECM). The signaling pathways that coordinate this complex communication, however, remain poorly understood. The adhesion G protein-coupled receptor ADGRG1 (also known as GPR56) is an evolutionarily conserved regulator of OL development in humans, mice, and zebrafish, although its activating ligand for OL lineage cells is unknown. Here, we report that microglia-derived transglutaminase-2 (TG2) signals to ADGRG1 on OL precursor cells (OPCs) in the presence of the ECM protein laminin and that TG2/laminin-dependent activation of ADGRG1 promotes OPC proliferation. Signaling by TG2/laminin to ADGRG1 on OPCs additionally improves remyelination in two murine models of demyelination. These findings identify a novel glia-to-glia signaling pathway that promotes myelin formation and repair, and suggest new strategies to enhance remyelination.
Collapse
Affiliation(s)
- Stefanie Giera
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Rong Luo
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Yanqin Ying
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States.,Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Sarah D Ackerman
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
| | - Sung-Jin Jeong
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neural Development and Diseases, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Hannah M Stoveken
- Department of Pharmacology, University of Michigan Medical Center, Ann Arbor, United States
| | - Christopher J Folts
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Christina A Welsh
- Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Gregory G Tall
- Department of Pharmacology, University of Michigan Medical Center, Ann Arbor, United States
| | - Beth Stevens
- Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
| | - Xianhua Piao
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, United States.,Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital and Harvard Medical School, Boston, United States
| |
Collapse
|
759
|
Abstract
Microglia are brain-resident macrophages whose function affects a myriad of physiological processes and can in turn be affected by peripheral factors. In a recent issue of Cell, Garel, Ginhoux and colleagues describe how gender, developmental stage, and microbiome contribute to the transcriptome of microglia (Thion et al., 2018).
Collapse
Affiliation(s)
- Antoine Louveau
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
760
|
Concepcion KR, Zhang L. Corticosteroids and perinatal hypoxic-ischemic brain injury. Drug Discov Today 2018; 23:1718-1732. [PMID: 29778695 DOI: 10.1016/j.drudis.2018.05.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/13/2018] [Accepted: 05/11/2018] [Indexed: 01/15/2023]
Abstract
Perinatal hypoxic-ischemic (HI) brain injury is the major cause of neonatal mortality and severe long-term neurological morbidity. Yet, the effective therapeutic interventions currently available are extremely limited. Corticosteroids act on both mineralocorticoid (MR) and glucocorticoid (GR) receptors and modulate inflammation and apoptosis in the brain. Neuroinflammatory response to acute cerebral HI is a major contributor to the pathophysiology of perinatal brain injury. Here, we give an overview of current knowledge of corticosteroid-mediated modulations of inflammation and apoptosis in the neonatal brain, focusing on key regulatory cells of the innate and adaptive immune response. In addition, we provide new insights into targets of MR and GR in potential therapeutic strategies that could be beneficial for the treatment of infants with HI brain injury.
Collapse
Affiliation(s)
- Katherine R Concepcion
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
761
|
Rothhammer V, Borucki DM, Tjon EC, Takenaka MC, Chao CC, Ardura-Fabregat A, de Lima KA, Gutiérrez-Vázquez C, Hewson P, Staszewski O, Blain M, Healy L, Neziraj T, Borio M, Wheeler M, Dragin LL, Laplaud DA, Antel J, Alvarez JI, Prinz M, Quintana FJ. Microglial control of astrocytes in response to microbial metabolites. Nature 2018; 557:724-728. [PMID: 29769726 DOI: 10.1038/s41586-018-0119-x] [Citation(s) in RCA: 727] [Impact Index Per Article: 103.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 04/11/2018] [Indexed: 12/14/2022]
Abstract
Microglia and astrocytes modulate inflammation and neurodegeneration in the central nervous system (CNS)1-3. Microglia modulate pro-inflammatory and neurotoxic activities in astrocytes, but the mechanisms involved are not completely understood4,5. Here we report that TGFα and VEGF-B produced by microglia regulate the pathogenic activities of astrocytes in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Microglia-derived TGFα acts via the ErbB1 receptor in astrocytes to limit their pathogenic activities and EAE development. Conversely, microglial VEGF-B triggers FLT-1 signalling in astrocytes and worsens EAE. VEGF-B and TGFα also participate in the microglial control of human astrocytes. Furthermore, expression of TGFα and VEGF-B in CD14+ cells correlates with the multiple sclerosis lesion stage. Finally, metabolites of dietary tryptophan produced by the commensal flora control microglial activation and TGFα and VEGF-B production, modulating the transcriptional program of astrocytes and CNS inflammation through a mechanism mediated by the aryl hydrocarbon receptor. In summary, we identified positive and negative regulators that mediate the microglial control of astrocytes. Moreover, these findings define a pathway through which microbial metabolites limit pathogenic activities of microglia and astrocytes, and suppress CNS inflammation. This pathway may guide new therapies for multiple sclerosis and other neurological disorders.
Collapse
Affiliation(s)
- Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Davis M Borucki
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily C Tjon
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maisa C Takenaka
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chun-Cheih Chao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Kalil Alves de Lima
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Gutiérrez-Vázquez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrick Hewson
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ori Staszewski
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Manon Blain
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Luke Healy
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Tradite Neziraj
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matilde Borio
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Loic Lionel Dragin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Jorge Ivan Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
762
|
CX3CR1 Does Not Universally Mediate Microglia-Neuron Crosstalk during Synaptic Plasticity. J Neurosci 2018; 38:4457-4459. [PMID: 29743344 DOI: 10.1523/jneurosci.3419-17.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 04/03/2018] [Accepted: 04/10/2018] [Indexed: 11/21/2022] Open
|
763
|
Abstract
Microglia differentiate from progenitors that infiltrate the nascent CNS during early embryonic development. They then remain in this unique immune-privileged environment throughout life. Multiple immune mechanisms, which we collectively refer to as microglial checkpoints, ensure efficient and tightly regulated microglial responses to perturbations in the CNS milieu. Such mechanisms are essential for proper CNS development and optimal physiological function. However, in chronic disease or aging, when a robust immune response is required, such checkpoint mechanisms may limit the ability of microglia to protect the CNS. Here we survey microglial checkpoint mechanisms and their roles in controlling microglial function throughout life and in disease, and discuss how they may be targeted therapeutically.
Collapse
|
764
|
Investigating the neuroimmunogenic architecture of schizophrenia. Mol Psychiatry 2018; 23:1251-1260. [PMID: 28485405 DOI: 10.1038/mp.2017.89] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/29/2017] [Accepted: 03/08/2017] [Indexed: 12/13/2022]
Abstract
The role of the immune system in schizophrenia remains controversial despite numerous studies to date. Most studies have profiled expression of select genes or proteins in peripheral blood, but none have focused on the expression of canonical pathways that mediate overall immune response. The current study used a systematic genetic approach to investigate the role of the immune system in a large sample of post-mortem brain of patients with schizophrenia: RNA sequencing was performed to assess the differential expression of 561 immune genes and 20 immune pathways in dorsolateral prefrontal cortex (DLPFC) (144 schizophrenia and 196 control subjects) and hippocampus (83 schizophrenia and 187 control subjects). The effect of RNA quality on gene expression was found to be highly correlated with the effect of diagnosis even after adjustment for observable RNA quality parameters (i.e. RNA integrity), thus this confounding relationship was statistically controlled using principal components derived from the gene expression matrix. In DLPFC, 23 immune genes were found to be differentially expressed (false discovery rate <0.05), of which seven genes replicated in both directionality and at nominal significance (P<0.05) in an independent post-mortem DLPFC data set (182 schizophrenia and 212 control subjects), although notably at least five of these genes have prominent roles in pathways other than immune function and overall the effect sizes were minimal (fold change <1.1). In the hippocampus, no individual immune genes were identified to be differentially expressed, and in both DLPFC and hippocampus none of the individual immune pathways were relatively differentially expressed. Further, genomic schizophrenia risk profiles scores were not correlated with the expression of individual immune pathways or differentially expressed genes. Overall, past reports claiming a primary pathogenic role of the immune system intrinsic to the brain in schizophrenia could not be confirmed.
Collapse
|
765
|
Report on the National Eye Institute's Audacious Goals Initiative: Creating a Cellular Environment for Neuroregeneration. eNeuro 2018; 5:eN-COM-0035-18. [PMID: 29766041 PMCID: PMC5952320 DOI: 10.1523/eneuro.0035-18.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 03/01/2018] [Accepted: 03/01/2018] [Indexed: 12/16/2022] Open
Abstract
The cellular environment of the CNS is non-permissive for growth and regeneration. In the retina, transplantation of stem cells has been limited by inefficient survival and integration into existing circuits. In November 2016, as part of the National Eye Institute's Audacious Goals Initiative (AGI), a diverse collection of investigators gathered for a workshop devoted to articulating the gaps in knowledge, barriers to progress, and ideas for new approaches to understanding cellular environments within the retina and how these environments may be manipulated. In doing so, the group identified the areas of (1) retinal and optic nerve glia, (2) microglia and inflammation, and the (3) extracellular matrix (ECM) and retinal vasculature as key to advancing our understanding and manipulation of the retinal microenvironments. We summarize here the findings of the workshop for the broader scientific community.
Collapse
|
766
|
Rosenberg AB, Roco CM, Muscat RA, Kuchina A, Sample P, Yao Z, Graybuck LT, Peeler DJ, Mukherjee S, Chen W, Pun SH, Sellers DL, Tasic B, Seelig G. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 2018; 360:176-182. [PMID: 29545511 PMCID: PMC7643870 DOI: 10.1126/science.aam8999] [Citation(s) in RCA: 895] [Impact Index Per Article: 127.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 09/30/2017] [Accepted: 02/26/2018] [Indexed: 12/11/2022]
Abstract
To facilitate scalable profiling of single cells, we developed split-pool ligation-based transcriptome sequencing (SPLiT-seq), a single-cell RNA-seq (scRNA-seq) method that labels the cellular origin of RNA through combinatorial barcoding. SPLiT-seq is compatible with fixed cells or nuclei, allows efficient sample multiplexing, and requires no customized equipment. We used SPLiT-seq to analyze 156,049 single-nucleus transcriptomes from postnatal day 2 and 11 mouse brains and spinal cords. More than 100 cell types were identified, with gene expression patterns corresponding to cellular function, regional specificity, and stage of differentiation. Pseudotime analysis revealed transcriptional programs driving four developmental lineages, providing a snapshot of early postnatal development in the murine central nervous system. SPLiT-seq provides a path toward comprehensive single-cell transcriptomic analysis of other similarly complex multicellular systems.
Collapse
Affiliation(s)
| | - Charles M Roco
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Richard A Muscat
- Department of Electrical Engineering, University of Washington, Seattle, WA, USA
| | - Anna Kuchina
- Department of Electrical Engineering, University of Washington, Seattle, WA, USA
| | - Paul Sample
- Department of Electrical Engineering, University of Washington, Seattle, WA, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - David J Peeler
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Sumit Mukherjee
- Department of Electrical Engineering, University of Washington, Seattle, WA, USA
| | - Wei Chen
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
| | - Suzie H Pun
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Drew L Sellers
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA
| | | | - Georg Seelig
- Department of Electrical Engineering, University of Washington, Seattle, WA, USA.
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
767
|
Lenz KM, Nelson LH. Microglia and Beyond: Innate Immune Cells As Regulators of Brain Development and Behavioral Function. Front Immunol 2018; 9:698. [PMID: 29706957 PMCID: PMC5908908 DOI: 10.3389/fimmu.2018.00698] [Citation(s) in RCA: 370] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/21/2018] [Indexed: 01/10/2023] Open
Abstract
Innate immune cells play a well-documented role in the etiology and disease course of many brain-based conditions, including multiple sclerosis, Alzheimer's disease, traumatic brain and spinal cord injury, and brain cancers. In contrast, it is only recently becoming clear that innate immune cells, primarily brain resident macrophages called microglia, are also key regulators of brain development. This review summarizes the current state of knowledge regarding microglia in brain development, with particular emphasis on how microglia during development are distinct from microglia later in life. We also summarize the effects of early life perturbations on microglia function in the developing brain, the role that biological sex plays in microglia function, and the potential role that microglia may play in developmental brain disorders. Finally, given how new the field of developmental neuroimmunology is, we highlight what has yet to be learned about how innate immune cells shape the development of brain and behavior.
Collapse
Affiliation(s)
- Kathryn M Lenz
- Department of Psychology, The Ohio State University, Columbus, OH, United States.,Department of Neuroscience, The Ohio State University, Columbus, OH, United States.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Lars H Nelson
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
768
|
Cronk JC, Filiano AJ, Louveau A, Marin I, Marsh R, Ji E, Goldman DH, Smirnov I, Geraci N, Acton S, Overall CC, Kipnis J. Peripherally derived macrophages can engraft the brain independent of irradiation and maintain an identity distinct from microglia. J Exp Med 2018; 215:1627-1647. [PMID: 29643186 PMCID: PMC5987928 DOI: 10.1084/jem.20180247] [Citation(s) in RCA: 301] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 11/04/2022] Open
Abstract
Peripherally derived macrophages infiltrate the brain after bone marrow transplantation and during central nervous system (CNS) inflammation. It was initially suggested that these engrafting cells were newly derived microglia and that irradiation was essential for engraftment to occur. However, it remains unclear whether brain-engrafting macrophages (beMφs) acquire a unique phenotype in the brain, whether long-term engraftment may occur without irradiation, and whether brain function is affected by the engrafted cells. In this study, we demonstrate that chronic, partial microglia depletion is sufficient for beMφs to populate the niche and that the presence of beMφs does not alter behavior. Furthermore, beMφs maintain a unique functional and transcriptional identity as compared with microglia. Overall, this study establishes beMφs as a unique CNS cell type and demonstrates that therapeutic engraftment of beMφs may be possible with irradiation-free conditioning regimens.
Collapse
Affiliation(s)
- James C Cronk
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA.,Department of Neuroscience, University of Virginia, Charlottesville, VA.,Graduate Program in Neuroscience, University of Virginia, Charlottesville, VA.,Medical Scientist Training Program, University of Virginia, Charlottesville, VA
| | - Anthony J Filiano
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA.,Department of Neuroscience, University of Virginia, Charlottesville, VA
| | - Antoine Louveau
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA.,Department of Neuroscience, University of Virginia, Charlottesville, VA
| | - Ioana Marin
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA.,Department of Neuroscience, University of Virginia, Charlottesville, VA.,Graduate Program in Neuroscience, University of Virginia, Charlottesville, VA
| | - Rachel Marsh
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA.,Department of Neuroscience, University of Virginia, Charlottesville, VA
| | - Emily Ji
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA.,Department of Neuroscience, University of Virginia, Charlottesville, VA
| | - Dylan H Goldman
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA.,Department of Neuroscience, University of Virginia, Charlottesville, VA.,Graduate Program in Neuroscience, University of Virginia, Charlottesville, VA
| | - Igor Smirnov
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA.,Department of Neuroscience, University of Virginia, Charlottesville, VA
| | - Nicholas Geraci
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA
| | - Scott Acton
- Virginia Image and Video Analysis Laboratory, Department of Electrical and Computer Engineering and Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Christopher C Overall
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA .,Department of Neuroscience, University of Virginia, Charlottesville, VA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA .,Department of Neuroscience, University of Virginia, Charlottesville, VA.,Graduate Program in Neuroscience, University of Virginia, Charlottesville, VA.,Medical Scientist Training Program, University of Virginia, Charlottesville, VA
| |
Collapse
|
769
|
Sevenich L. Brain-Resident Microglia and Blood-Borne Macrophages Orchestrate Central Nervous System Inflammation in Neurodegenerative Disorders and Brain Cancer. Front Immunol 2018; 9:697. [PMID: 29681904 PMCID: PMC5897444 DOI: 10.3389/fimmu.2018.00697] [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: 01/31/2018] [Accepted: 03/21/2018] [Indexed: 01/09/2023] Open
Abstract
Inflammation is a hallmark of different central nervous system (CNS) pathologies. It has been linked to neurodegenerative disorders as well as primary and metastatic brain tumors. Microglia, the brain-resident immune cells, are emerging as a central player in regulating key pathways in CNS inflammation. Recent insights into neuroinflammation indicate that blood-borne immune cells represent an additional critical cellular component in mediating CNS inflammation. The lack of experimental systems that allow for discrimination between brain-resident and recruited myeloid cells has previously halted functional analysis of microglia and their blood-borne counterparts in brain malignancies. However, recent conceptual and technological advances, such as the generation of lineage tracing models and the identification of cell type-specific markers provide unprecedented opportunities to study the cellular functions of microglia and macrophages by functional interference. The use of different “omic” strategies as well as imaging techniques has significantly increased our knowledge of disease-associated gene signatures and effector functions under pathological conditions. In this review, recent developments in evaluating functions of brain-resident and recruited myeloid cells in neurodegenerative disorders and brain cancers will be discussed and unique or shared cellular traits of microglia and macrophages in different CNS disorders will be highlighted. Insight from these studies will shape our understanding of disease- and cell-type-specific effector functions of microglia or macrophages and will open new avenues for therapeutic intervention that target aberrant functions of myeloid cells in CNS pathologies.
Collapse
Affiliation(s)
- Lisa Sevenich
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| |
Collapse
|
770
|
Testicular macrophages: Guardians of fertility. Cell Immunol 2018; 330:120-125. [PMID: 29650243 DOI: 10.1016/j.cellimm.2018.03.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 03/22/2018] [Accepted: 03/29/2018] [Indexed: 12/23/2022]
Abstract
Macrophages are innate immune cells present in essentially every organ of the body with dedicated tissue specific functions. We will present in this review the unique properties and functions of macrophage populations residing in the testis, an immune-privileged organ. Testicular macrophages (tMΦ) could be seen as guardians of fertility due to their immunosuppressive functions protecting spermatogenesis from auto immune-attack. They exhibit testis specific functions with essential roles in normal testis homeostasis and fetal testicular development. Recently, two distinct testicular macrophage populations have been characterized based on different localization, morphology, gene expression profiles, developmental origin and postnatal development. We will discuss the importance of these two testicular macrophage populations for organ specific functions such as testosterone production and spermatogenesis, as well as their role in establishing immuno-privilege highlighting the contributions of macrophages to male fertility.
Collapse
|
771
|
Wilhelmsson U, Andersson D, de Pablo Y, Pekny R, Ståhlberg A, Mulder J, Mitsios N, Hortobágyi T, Pekny M, Pekna M. Injury Leads to the Appearance of Cells with Characteristics of Both Microglia and Astrocytes in Mouse and Human Brain. Cereb Cortex 2018; 27:3360-3377. [PMID: 28398520 DOI: 10.1093/cercor/bhx069] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Indexed: 12/21/2022] Open
Abstract
Microglia and astrocytes have been considered until now as cells with very distinct identities. Here, we assessed the heterogeneity within microglia/monocyte cell population in mouse hippocampus and determined their response to injury, by using single-cell gene expression profiling of cells isolated from uninjured and deafferented hippocampus. We found that in individual cells, microglial markers Cx3cr1, Aif1, Itgam, and Cd68 were co-expressed. Interestingly, injury led to the co-expression of the astrocyte marker Gfap in a subpopulation of Cx3cr1-expressing cells from both the injured and contralesional hippocampus. Cells co-expressing astrocyte and microglia markers were also detected in the in vitro LPS activation/injury model and in sections from human brain affected by stroke, Alzheimer's disease, and Lewy body dementia. Our findings indicate that injury and chronic neurodegeneration lead to the appearance of cells that share molecular characteristics of both microglia and astrocytes, 2 cell types with distinct embryologic origin and function.
Collapse
Affiliation(s)
- Ulrika Wilhelmsson
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Daniel Andersson
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Yolanda de Pablo
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Roy Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Anders Ståhlberg
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Jan Mulder
- Science for Life Laboratory, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas Mitsios
- Science for Life Laboratory, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tibor Hortobágyi
- Division of Neuropathology, Institute of Pathology, Faculty of Medicine, University of Debrecen, Hungary.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden.,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| | - Marcela Pekna
- Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 405 30 Gothenburg, Sweden.,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| |
Collapse
|
772
|
Lu Y, Zhang XS, Zhang ZH, Zhou XM, Gao YY, Liu GJ, Wang H, Wu LY, Li W, Hang CH. Peroxiredoxin 2 activates microglia by interacting with Toll-like receptor 4 after subarachnoid hemorrhage. J Neuroinflammation 2018; 15:87. [PMID: 29554978 PMCID: PMC5859544 DOI: 10.1186/s12974-018-1118-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/06/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Peroxiredoxin (Prx) protein family have been reported as important damage-associated molecular patterns (DAMPs) in ischemic stroke. Since peroxiredoxin 2 (Prx2) is the third most abundant protein in erythrocytes and the second most protein in the cerebrospinal fluid in traumatic brain injury and subarachnoid hemorrhage (SAH) patients, we assessed the role of extracellular Prx2 in the context of SAH. METHODS We introduced a co-culture system of primary neurons and microglia. Prx2 was added to culture medium with oxyhemoglobin (OxyHb) to mimic SAH in vitro. Neuronal cell viability was assessed by lactate dehydrogenase (LDH) assay, and neuronal apoptosis was determined by TUNEL staining. Inflammatory factors in culture medium were measured by ELISA, and their mRNA levels in microglia were determined by qPCR. Toll-like receptor 4 knockout (TLR4-KO) mice were used to provide TLR4-KO microglia; ST-2825 was used to inhibit MyD88, and pyrrolidine dithiocarbamate (PDTC) was used to inhibit NF-κB. Related cellular signals were analyzed by Western blot. Furthermore, we detected the level of Prx2 in aneurysmal SAH patients' cerebrospinal fluids (CSF) and compared its relationship with Hunt-Hess grades. RESULTS Prx2 interacted with TLR4 on microglia after SAH and then activated microglia through TLR4/MyD88/NF-κB signaling pathway. Pro-inflammatory factors were expressed and released, eventually caused neuronal apoptosis. The levels of Prx2 in SAH patients positively correlated with Hunt-Hess grades. CONCLUSIONS Extracellular Prx2 in CSF after SAH is a DAMP which resulted in microglial activation via TLR4/MyD88/NF-κB pathway and then neuronal apoptosis. Prx2 in patients' CSF may be a potential indicator of brain injury and prognosis.
Collapse
Affiliation(s)
- Yue Lu
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiang-Sheng Zhang
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zi-Huan Zhang
- Department of Neurosurgery, Zhongdu Hospital, Bengbu, China
| | - Xiao-Ming Zhou
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yong-Yue Gao
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guang-Jie Liu
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Han Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, South Medical University, Nanjing, China
| | - Ling-Yun Wu
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wei Li
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Chun-Hua Hang
- Department of Neurosurgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
773
|
Ding J, Hagood JS, Ambalavanan N, Kaminski N, Bar-Joseph Z. iDREM: Interactive visualization of dynamic regulatory networks. PLoS Comput Biol 2018. [PMID: 29538379 PMCID: PMC5868853 DOI: 10.1371/journal.pcbi.1006019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Dynamic Regulatory Events Miner (DREM) software reconstructs dynamic regulatory networks by integrating static protein-DNA interaction data with time series gene expression data. In recent years, several additional types of high-throughput time series data have been profiled when studying biological processes including time series miRNA expression, proteomics, epigenomics and single cell RNA-Seq. Combining all available time series and static datasets in a unified model remains an important challenge and goal. To address this challenge we have developed a new version of DREM termed interactive DREM (iDREM). iDREM provides support for all data types mentioned above and combines them with existing interaction data to reconstruct networks that can lead to novel hypotheses on the function and timing of regulators. Users can interactively visualize and query the resulting model. We showcase the functionality of the new tool by applying it to microglia developmental data from multiple labs.
Collapse
Affiliation(s)
- Jun Ding
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - James S. Hagood
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego and Rady Children’s Hospital, La Jolla, California, United States of America
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Ziv Bar-Joseph
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
774
|
Antonson AM, Balakrishnan B, Radlowski EC, Petr G, Johnson RW. Altered Hippocampal Gene Expression and Morphology in Fetal Piglets following Maternal Respiratory Viral Infection. Dev Neurosci 2018. [PMID: 29539630 DOI: 10.1159/000486850] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Maternal infection during pregnancy increases the risk of neurobehavioral problems in offspring. Evidence from rodent models indicates that the maternal immune response to infection can alter fetal brain development, particularly in the hippocampus. However, information on the effects of maternal viral infection on fetal brain development in gyrencephalic species is limited. Thus, the objective of this study was to assess several effects of maternal viral infection in the last one-third of gestation on hippocampal gene expression and development in fetal piglets. Pregnant gilts were inoculated with porcine reproductive and respiratory syndrome virus (PRRSV) at gestational day (GD) 76 and the fetuses were removed by cesarean section at GD 111 (3 days before anticipated parturition). The gilts infected with PRRSV had elevated plasma interleukin-6 levels and developed transient febrile and anorectic responses lasting approximately 21 days. Despite having a similar overall body weight, fetuses from the PRRSV-infected gilts had a decreased brain weight and altered hippocampal gene expression compared to fetuses from control gilts. Notably, maternal infection caused a reduction in estimated neuronal numbers in the fetal dentate gyrus and subiculum. The number of proliferative Ki-67+ cells was not altered, but the relative integrated density of GFAP+ staining was increased, in addition to an increase in GFAP gene expression, indicating astrocyte-specific gliosis. Maternal viral infection caused an increase in fetal hippocampal gene expression of the inflammatory cytokines TNF-α and IFN-γ and the myelination marker myelin basic protein. MHCII protein, a classic monocyte activation marker, was reduced in microglia, while expression of the MHCII gene was decreased in hippocampal tissue of the fetuses from PRRSV-infected gilts. Together, these data suggest that maternal viral infection at the beginning of the last trimester results in a reduction in fetal hippocampal neurons that is evident 5 weeks after infection, when fetal piglets are near full term. The neuronal reduction was not accompanied by pronounced neuroinflammation at GD 111, indicating that any activation of classic neuroinflammatory pathways by maternal viral infection, if present, is mostly resolved by parturition.
Collapse
Affiliation(s)
- Adrienne M Antonson
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Bindu Balakrishnan
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Emily C Radlowski
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Geraldine Petr
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Rodney W Johnson
- Department of Animal Sciences, Laboratory of Integrative Immunology and Behavior, University of Illinois Urbana-Champaign, Urbana, Illinois, USA.,Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA.,Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
775
|
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that develops insidiously and causes dementia finally. There are also clinical complications in advanced dementia, such as eating problems, infections, which will lead to the decline of patients' life quality, and the rising cost of care for AD to our society. AD will be important public health challenge. Early detection of AD may be a key issue to prevent, delay, and stop the disease. Gut microbiome and neuroinflammation are closely related with nervous system diseases, although the specific mechanism is not clear. This review introduces the relationship between neuroinflammation, gut microbiome, and AD.
Collapse
|
776
|
Cristiano C, Lama A, Lembo F, Mollica MP, Calignano A, Mattace Raso G. Interplay Between Peripheral and Central Inflammation in Autism Spectrum Disorders: Possible Nutritional and Therapeutic Strategies. Front Physiol 2018; 9:184. [PMID: 29563885 PMCID: PMC5845898 DOI: 10.3389/fphys.2018.00184] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/20/2018] [Indexed: 12/16/2022] Open
Abstract
Pre- and post-natal factors can affect brain development and function, impacting health outcomes with particular relevance to neurodevelopmental diseases, such as autism spectrum disorders (ASDs). Maternal obesity and its associated complications have been related to the increased risk of ASDs in offspring. Indeed, animals exposed to maternal obesity or high fat diets are prone to social communication impairment and repetitive behavior, the hallmarks of autism. During development, fatty acids and sugars, as well as satiety hormones, like insulin and leptin, and inflammatory factors related to obesity-induced low grade inflammation, could play a role in the impairment of neuroendocrine system and brain neuronal circuits regulating behavior in offspring. On the other side, post-natal factors, such as mode of delivery, stress, diet, or antibiotic treatment are associated to a modification of gut microbiota composition, perturbing microbiota-gut-brain axis. Indeed, the interplay between the gastrointestinal tract and the central nervous system not only occurs through neural, hormonal, and immune pathways, but also through microbe-derived metabolic products. The modification of unhealthy perinatal and postnatal environment, manipulation of gut microbiota, nutritional, and dietary interventions could represent possible strategies in preventing or limiting ASDs, through targeting inflammatory process and gut microbiota.
Collapse
Affiliation(s)
- Claudia Cristiano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Francesca Lembo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Maria P Mollica
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
777
|
Cloarec R, Bauer S, Teissier N, Schaller F, Luche H, Courtens S, Salmi M, Pauly V, Bois E, Pallesi-Pocachard E, Buhler E, Michel FJ, Gressens P, Malissen M, Stamminger T, Streblow DN, Bruneau N, Szepetowski P. In Utero Administration of Drugs Targeting Microglia Improves the Neurodevelopmental Outcome Following Cytomegalovirus Infection of the Rat Fetal Brain. Front Cell Neurosci 2018; 12:55. [PMID: 29559892 PMCID: PMC5845535 DOI: 10.3389/fncel.2018.00055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/16/2018] [Indexed: 01/20/2023] Open
Abstract
Congenital cytomegalovirus (CMV) infections represent one leading cause of neurodevelopmental disorders. Recently, we reported on a rat model of CMV infection of the developing brain in utero, characterized by early and prominent infection and alteration of microglia-the brain-resident mononuclear phagocytes. Besides their canonical function against pathogens, microglia are also pivotal to brain development. Here we show that CMV infection of the rat fetal brain recapitulated key postnatal phenotypes of human congenital CMV including increased mortality, sensorimotor impairment reminiscent of cerebral palsy, hearing defects, and epileptic seizures. The possible influence of early microglia alteration on those phenotypes was then questioned by pharmacological targeting of microglia during pregnancy. One single administration of clodronate liposomes in the embryonic brains at the time of CMV injection to deplete microglia, and maternal feeding with doxycyxline throughout pregnancy to modify microglia in the litters' brains, were both associated with dramatic improvements of survival, body weight gain, sensorimotor development and with decreased risk of epileptic seizures. Improvement of microglia activation status did not persist postnatally after doxycycline discontinuation; also, active brain infection remained unchanged by doxycycline. Altogether our data indicate that early microglia alteration, rather than brain CMV load per se, is instrumental in influencing survival and the neurological outcomes of CMV-infected rats, and suggest that microglia might participate in the neurological outcome of congenital CMV in humans. Furthermore this study represents a first proof-of-principle for the design of microglia-targeted preventive strategies in the context of congenital CMV infection of the brain.
Collapse
Affiliation(s)
- Robin Cloarec
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France.,Neurochlore, Marseille, France
| | - Sylvian Bauer
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France
| | - Natacha Teissier
- French National Institute of Health and Medical Research INSERM U1141, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,PremUP, Paris, France
| | - Fabienne Schaller
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France.,PPGI Platform, INMED, Marseille, France
| | - Hervé Luche
- Centre National de la Recherche Scientifique CNRS UMS3367, CIPHE (Centre D'Immunophénomique), French National Institute of Health and Medical Research INSERM US012, PHENOMIN, Aix-Marseille University, Marseille, France
| | - Sandra Courtens
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France
| | - Manal Salmi
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France
| | - Vanessa Pauly
- Laboratoire de Santé Publique EA 3279, Faculté de Médecine Centre d'Evaluation de la Pharmacodépendance-Addictovigilance de Marseille (PACA-Corse) Associé, Aix-Marseille University, Marseille, France
| | - Emilie Bois
- French National Institute of Health and Medical Research INSERM U1141, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,PremUP, Paris, France
| | - Emilie Pallesi-Pocachard
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France.,PBMC platform, INMED, Marseille, France
| | - Emmanuelle Buhler
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France.,PPGI Platform, INMED, Marseille, France
| | - François J Michel
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France.,InMAGIC platform, INMED, Marseille, France
| | - Pierre Gressens
- French National Institute of Health and Medical Research INSERM U1141, Paris Diderot University, Sorbonne Paris Cité, Paris, France.,PremUP, Paris, France
| | - Marie Malissen
- Centre National de la Recherche Scientifique CNRS UMS3367, CIPHE (Centre D'Immunophénomique), French National Institute of Health and Medical Research INSERM US012, PHENOMIN, Aix-Marseille University, Marseille, France
| | - Thomas Stamminger
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Nadine Bruneau
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France
| | - Pierre Szepetowski
- INMED, French National Institute of Health and Medical Research INSERM U1249, Aix-Marseille University, Marseille, France
| |
Collapse
|
778
|
A story of birth and death: Insights into the formation and dynamics of the microglial population. Brain Behav Immun 2018; 69:9-17. [PMID: 28341583 DOI: 10.1016/j.bbi.2017.03.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/16/2017] [Accepted: 03/20/2017] [Indexed: 12/24/2022] Open
Abstract
Microglia are the main resident immunocompetent cells of the brain with key roles in brain development, homeostasis and function. Here we briefly review our current knowledge of the homeostatic mechanisms regulating the composition and turnover of the microglial population under physiological conditions from development to ageing. A greater understanding of these mechanisms may inform understanding of how dysregulation of microglial dynamics could contribute to the pathogenesis and/or progression of neurological disorders.
Collapse
|
779
|
Ciernia AV, Careaga M, Ashwood P, LaSalle J. Microglia from offspring of dams with allergic asthma exhibit epigenomic alterations in genes dysregulated in autism. Glia 2018; 66:505-521. [PMID: 29134693 PMCID: PMC5767155 DOI: 10.1002/glia.23261] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/18/2017] [Accepted: 10/25/2017] [Indexed: 12/24/2022]
Abstract
Dysregulation in immune responses during pregnancy increases the risk of a having a child with an autism spectrum disorder (ASD). Asthma is one of the most common chronic diseases among pregnant women, and symptoms often worsen during pregnancy. We recently developed a mouse model of maternal allergic asthma (MAA) that induces changes in sociability, repetitive, and perseverative behaviors in the offspring. Since epigenetic changes help a static genome adapt to the maternal environment, activation of the immune system may epigenetically alter fetal microglia, the brain's resident immune cells. We therefore tested the hypothesis that epigenomic alterations to microglia may be involved in behavioral abnormalities observed in MAA offspring. We used the genome-wide approaches of whole genome bisulfite sequencing to examine DNA methylation and RNA sequencing to examine gene expression in microglia from juvenile MAA offspring. Differentially methylated regions were enriched for immune signaling pathways and important microglial developmental transcription factor binding motifs. Differential expression analysis identified genes involved in controlling microglial sensitivity to the environment and shaping neuronal connections in the developing brain. Differentially expressed genes significantly overlapped genes with altered expression in human ASD cortex, supporting a role for microglia in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Annie Vogel Ciernia
- Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616
| | - Milo Careaga
- MIND Institute, 2825 50 Street, Sacramento, CA 95817, University of California, Davis
| | - Paul Ashwood
- MIND Institute, 2825 50 Street, Sacramento, CA 95817, University of California, Davis
| | - Janine LaSalle
- Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616
| |
Collapse
|
780
|
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: 55] [Impact Index Per Article: 7.9] [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
|
781
|
Single-nucleus analysis of accessible chromatin in developing mouse forebrain reveals cell-type-specific transcriptional regulation. Nat Neurosci 2018; 21:432-439. [PMID: 29434377 DOI: 10.1038/s41593-018-0079-3] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/27/2017] [Indexed: 01/23/2023]
Abstract
Analysis of chromatin accessibility can reveal transcriptional regulatory sequences, but heterogeneity of primary tissues poses a significant challenge in mapping the precise chromatin landscape in specific cell types. Here we report single-nucleus ATAC-seq, a combinatorial barcoding-assisted single-cell assay for transposase-accessible chromatin that is optimized for use on flash-frozen primary tissue samples. We apply this technique to the mouse forebrain through eight developmental stages. Through analysis of more than 15,000 nuclei, we identify 20 distinct cell populations corresponding to major neuronal and non-neuronal cell types. We further define cell-type-specific transcriptional regulatory sequences, infer potential master transcriptional regulators and delineate developmental changes in forebrain cellular composition. Our results provide insight into the molecular and cellular dynamics that underlie forebrain development in the mouse and establish technical and analytical frameworks that are broadly applicable to other heterogeneous tissues.
Collapse
|
782
|
Abstract
High-throughput sequencing assays have become an increasingly common part of biological research across multiple fields. Even as the resulting sequences pile up in public databases, it is not always obvious how to make use of these data sets. Functional genomics offers approaches to integrate these "big" data into our understanding of rheumatic diseases. This review aims to provide a primer on thinking about big data from functional genomics in the context of rheumatology, using examples from the field's literature as well as the author's own work to illustrate the execution of functional genomics research. Study design is crucial to ensure the right samples are used to address the question of interest. In addition, sequencing assays produce a variety of data types, from gene expression to 3D chromatin structure and single-cell technologies, that can be integrated into a model of the underlying gene regulatory networks. The best approach for this analysis uses the scientific process: bioinformatic methods should be used in an iterative, hypothesis-driven manner to uncover the disease mechanism. Finally, the future of functional genomics will see big data fully integrated into rheumatology, leading to computationally trained researchers and interactive databases. The goal of this review is not to provide a manual, but to enhance the familiarity of readers with functional genomic approaches and provide a better sense of the challenges and possibilities.
Collapse
Affiliation(s)
- Deborah R Winter
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
783
|
Mukherjee D, Ignatowska-Jankowska BM, Itskovits E, Gonzales BJ, Turm H, Izakson L, Haritan D, Bleistein N, Cohen C, Amit I, Shay T, Grueter B, Zaslaver A, Citri A. Salient experiences are represented by unique transcriptional signatures in the mouse brain. eLife 2018; 7:e31220. [PMID: 29412137 PMCID: PMC5862526 DOI: 10.7554/elife.31220] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/05/2018] [Indexed: 12/11/2022] Open
Abstract
It is well established that inducible transcription is essential for the consolidation of salient experiences into long-term memory. However, whether inducible transcription relays information about the identity and affective attributes of the experience being encoded, has not been explored. To this end, we analyzed transcription induced by a variety of rewarding and aversive experiences, across multiple brain regions. Our results describe the existence of robust transcriptional signatures uniquely representing distinct experiences, enabling near-perfect decoding of recent experiences. Furthermore, experiences with shared attributes display commonalities in their transcriptional signatures, exemplified in the representation of valence, habituation and reinforcement. This study introduces the concept of a neural transcriptional code, which represents the encoding of experiences in the mouse brain. This code is comprised of distinct transcriptional signatures that correlate to attributes of the experiences that are being committed to long-term memory.
Collapse
Affiliation(s)
- Diptendu Mukherjee
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | | | - Eyal Itskovits
- Department of Genetics, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
- School of Computer Science and EngineeringThe Hebrew UniversityJerusalemIsrael
| | - Ben Jerry Gonzales
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Hagit Turm
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Liz Izakson
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Doron Haritan
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Noa Bleistein
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Chen Cohen
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Ido Amit
- Department of ImmunologyWeizmann Institute of ScienceRehovotIsrael
| | - Tal Shay
- Department of Life SciencesBen-Gurion University of the NegevBeer-ShevaIsrael
| | - Brad Grueter
- Department of PsychiatryVanderbilt University School of MedicineNashvilleUnited States
| | - Alon Zaslaver
- Department of Genetics, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Ami Citri
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
- The Edmond and Lily Safra Center for Brain SciencesThe Hebrew UniversityJerusalemIsrael
- Child and Brain Development ProgramCanadian Institute for Advanced ResearchTorontoCanada
| |
Collapse
|
784
|
Hirbec H, Marmai C, Duroux-Richard I, Roubert C, Esclangon A, Croze S, Lachuer J, Peyroutou R, Rassendren F. The microglial reaction signature revealed by RNAseq from individual mice. Glia 2018; 66:971-986. [PMID: 29399880 DOI: 10.1002/glia.23295] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/23/2017] [Accepted: 01/02/2018] [Indexed: 12/20/2022]
Abstract
Microglial cells have a double life as the immune cells of the brain in times of stress but have also specific physiological functions in homeostatic conditions. In pathological contexts, microglia undergo a phenotypic switch called "reaction" that promotes the initiation and the propagation of neuro-inflammation. Reaction is complex, molecularly heterogeneous and still poorly characterized, leading to the concept that microglial reactivity might be too diverse to be molecularly defined. However, it remains unknown whether reactive microglia from different pathological contexts share a common molecular signature. Using improved flow cytometry and RNAseq approaches we studied, with higher statistical power, the remodeling of microglia transcriptome in a mouse model of sepsis. Through bioinformatic comparison of our results with published datasets, we defined the microglial reactome as a set of genes discriminating reactive from homeostatic microglia. Ultimately, we identified a subset of 86 genes deregulated in both acute and neurodegenerative conditions. Our data provide a new comprehensive resource that includes functional analysis and specific molecular markers of microglial reaction which represent new tools for its unambiguous characterization.
Collapse
Affiliation(s)
- Hélène Hirbec
- Institute of Functional Genomics, CNRS, INSERM, University of Montpellier, Montpellier, France.,Labex ICST, Montpellier, France
| | - Camille Marmai
- Institute of Functional Genomics, CNRS, INSERM, University of Montpellier, Montpellier, France.,Labex ICST, Montpellier, France
| | | | | | | | - Séverine Croze
- ProfileXpert, Université Claude Bernard Lyon 1, Lyon, France
| | - Joël Lachuer
- ProfileXpert, Université Claude Bernard Lyon 1, Lyon, France
| | - Ronan Peyroutou
- Institute of Functional Genomics, CNRS, INSERM, University of Montpellier, Montpellier, France.,Labex ICST, Montpellier, France
| | - François Rassendren
- Institute of Functional Genomics, CNRS, INSERM, University of Montpellier, Montpellier, France.,Labex ICST, Montpellier, France
| |
Collapse
|
785
|
Claes C, Van den Daele J, Verfaillie CM. Generating tissue-resident macrophages from pluripotent stem cells: Lessons learned from microglia. Cell Immunol 2018; 330:60-67. [PMID: 29433896 DOI: 10.1016/j.cellimm.2018.01.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/31/2018] [Accepted: 01/31/2018] [Indexed: 02/07/2023]
Abstract
Over the past decades, the importance of the immune system in a broad scope of pathologies, has drawn attention towards tissue-resident macrophages, such as microglia in the brain. To enable the study of for instance microglia, it is crucial to recreate in vitro (and in vivo) assays. However, very fast loss of tissue-specific features of primary tissue resident macrophages, including microglia, upon in vitro culture has complicated such studies. Moreover, limited knowledge of macrophage developmental pathways and the role of local 'niche factors', has hampered the generation of tissue-resident macrophages from pluripotent stem cells (PSC). Recent data on the ontogeny of tissue-resident macrophages, combined with bulk and single cell RNAseq studies have identified the distinct origins and gene profile of microglia compared to other myeloid cells. As a result, over the past years, protocols have been published to create hPSC-derived microglia-'like' cells, as these cells are considered potential new therapeutic targets for therapies to treat neurodegenerative diseases. In this review we will provide an overview of different approaches taken to generate human microglia in vitro, taking into account their origin, and resemblance to their in vivo counterpart. Finally, we will discuss cell-extrinsic (culture conditions) and intrinsic factors (transcriptional machinery and epigenetics) that we believe can improve future differentiation protocols of tissue-resident macrophages from stem cells.
Collapse
Affiliation(s)
- Christel Claes
- Department of Development and Regeneration, Stem Cell and Developmental Biology, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Johanna Van den Daele
- Department of Development and Regeneration, Stem Cell and Developmental Biology, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Stem Cell and Developmental Biology, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium.
| |
Collapse
|
786
|
Prins JR, Eskandar S, Eggen BJL, Scherjon SA. Microglia, the missing link in maternal immune activation and fetal neurodevelopment; and a possible link in preeclampsia and disturbed neurodevelopment? J Reprod Immunol 2018; 126:18-22. [PMID: 29421625 DOI: 10.1016/j.jri.2018.01.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/06/2017] [Accepted: 01/17/2018] [Indexed: 12/24/2022]
Abstract
Disturbances in fetal neurodevelopment have extensively been related to neurodevelopmental disorders in early and later life. Fetal neurodevelopment is dependent on adequate functioning of the fetal immune system. During pregnancy, the maternal immune system is challenged to both tolerate the semi-allogenic fetus and to protect the mother and fetus from microbes. The fetal immune system is influenced by maternal immune disturbances; therefore, perturbations in maternal immunity likely do not only alter pregnancy outcome but also alter fetal neurodevelopment. A possible common pathway could be modulating the functioning of tissue macrophages in the placenta and brain. Maternal immune tolerance towards the fetus involves several complex adaptations. In this active maternal immune state, the fetus develops its own immunity. As cytokines and other players of the immune system -which can pass the placenta- are involved in neurodevelopment, disruptions in immune balance influence fetal neurodevelopment. Several studies reported an association between maternal immune activation, complications of pregnancy as preeclampsia, and altered neonatal neurodevelopment. A possible pathway involves dysfunctioning of microglia cells, the immune cells of the brain. Functionality of microglia cells during normal pregnancy is, however, poorly understood. The recent outbreak of ZIKA virus (ZKV), but also the literature on virus infections in general and its consequences on microglial cell function and fetal neurodevelopment show the devastating effects a virus infection during pregnancy can have.
Collapse
Affiliation(s)
- Jelmer R Prins
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands; Department of Obstetrics and Gynecology, Medisch Spectrum Twente, PO Box 50 000, 7500 KA Enschede, The Netherlands.
| | - Sharon Eskandar
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands; Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands.
| | - Bart J L Eggen
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands.
| | - Sicco A Scherjon
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
787
|
Korin B, Dubovik T, Rolls A. Mass cytometry analysis of immune cells in the brain. Nat Protoc 2018; 13:377-391. [DOI: 10.1038/nprot.2017.155] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
788
|
Bove RM. Why monkeys do not get multiple sclerosis (spontaneously): An evolutionary approach. EVOLUTION MEDICINE AND PUBLIC HEALTH 2018; 2018:43-59. [PMID: 29492266 PMCID: PMC5824939 DOI: 10.1093/emph/eoy002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022]
Abstract
The goal of this review is to apply an evolutionary lens to understanding the origins of multiple sclerosis (MS), integrating three broad observations. First, only humans are known to develop MS spontaneously. Second, humans have evolved large brains, with characteristically large amounts of metabolically costly myelin. This myelin is generated over long periods of neurologic development—and peak MS onset coincides with the end of myelination. Third, over the past century there has been a disproportionate increase in the rate of MS in young women of childbearing age, paralleling increasing westernization and urbanization, indicating sexually specific susceptibility in response to changing exposures. From these three observations about MS, a life history approach leads us to hypothesize that MS arises in humans from disruption of the normal homeostatic mechanisms of myelin production and maintenance, during our uniquely long myelination period. This review will highlight under-explored areas of homeostasis in brain development, that are likely to shed new light on the origins of MS and to raise further questions about the interactions between our ancestral genes and modern environments.
Collapse
Affiliation(s)
- Riley M Bove
- Department of Neurology, UCSF, San Francisco, CA, USA
| |
Collapse
|
789
|
Mecca C, Giambanco I, Donato R, Arcuri C. Microglia and Aging: The Role of the TREM2-DAP12 and CX3CL1-CX3CR1 Axes. Int J Mol Sci 2018; 19:E318. [PMID: 29361745 PMCID: PMC5796261 DOI: 10.3390/ijms19010318] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/21/2022] Open
Abstract
Depending on the species, microglial cells represent 5-20% of glial cells in the adult brain. As the innate immune effector of the brain, microglia are involved in several functions: regulation of inflammation, synaptic connectivity, programmed cell death, wiring and circuitry formation, phagocytosis of cell debris, and synaptic pruning and sculpting of postnatal neural circuits. Moreover, microglia contribute to some neurodevelopmental disorders such as Nasu-Hakola disease (NHD), and to aged-associated neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and others. There is evidence that human and rodent microglia may become senescent. This event determines alterations in the microglia activation status, associated with a chronic inflammation phenotype and with the loss of neuroprotective functions that lead to a greater susceptibility to the neurodegenerative diseases of aging. In the central nervous system (CNS), Triggering Receptor Expressed on Myeloid Cells 2-DNAX activation protein 12 (TREM2-DAP12) is a signaling complex expressed exclusively in microglia. As a microglial surface receptor, TREM2 interacts with DAP12 to initiate signal transduction pathways that promote microglial cell activation, phagocytosis, and microglial cell survival. Defective TREM2-DAP12 functions play a central role in the pathogenesis of several diseases. The CX3CL1 (fractalkine)-CX3CR1 signaling represents the most important communication channel between neurons and microglia. The expression of CX3CL1 in neurons and of its receptor CX3CR1 in microglia determines a specific interaction, playing fundamental roles in the regulation of the maturation and function of these cells. Here, we review the role of the TREM2-DAP12 and CX3CL1-CX3CR1 axes in aged microglia and the involvement of these pathways in physiological CNS aging and in age-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Carmen Mecca
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Ileana Giambanco
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Rosario Donato
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
- Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Cataldo Arcuri
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| |
Collapse
|
790
|
Winkler CW, Peterson KE. Using immunocompromised mice to identify mechanisms of Zika virus transmission and pathogenesis. Immunology 2018; 153:443-454. [PMID: 29266213 DOI: 10.1111/imm.12883] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/13/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV) is responsible for a recent global epidemic that has been associated with congenital brain malformations in fetuses and with Guillain-Barré syndrome in adults. Within the last 2 years, a major effort has been made to develop murine models to study the mechanism of viral transmission, pathogenesis and the host immune response. Here, we discuss the findings from these models regarding the role that the innate and adaptive immune responses have in controlling ZIKV infection and pathogenesis. Additionally, we examine how innate and adaptive immune responses influence sexual and vertical transmission of ZIKV infection as well as how these responses can influence the ability of ZIKV to cross the placenta and to induce damage in the developing brain.
Collapse
Affiliation(s)
- Clayton W Winkler
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Karin E Peterson
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| |
Collapse
|
791
|
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: 40] [Impact Index Per Article: 5.7] [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
|
792
|
Cell-specific and region-specific transcriptomics in the multiple sclerosis model: Focus on astrocytes. Proc Natl Acad Sci U S A 2018; 115:E302-E309. [PMID: 29279367 PMCID: PMC5777065 DOI: 10.1073/pnas.1716032115] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Changes in gene expression that occur across the central nervous system (CNS) during neurological diseases do not address the heterogeneity of cell types from one CNS region to another and are complicated by alterations in cellular composition during disease. Multiple sclerosis (MS) is multifocal by definition. Here, a cell-specific and region-specific transcriptomics approach was used to determine gene expression changes in astrocytes in the most widely used MS model, experimental autoimmune encephalomyelitis (EAE). Astrocyte-specific RNAs from various neuroanatomic regions were attained using RiboTag technology. Sequencing and bioinformatics analyses showed that EAE-induced gene expression changes differed between neuroanatomic regions when comparing astrocytes from spinal cord, cerebellum, cerebral cortex, and hippocampus. The top gene pathways that were changed in astrocytes from spinal cord during chronic EAE involved decreases in expression of cholesterol synthesis genes while immune pathway gene expression in astrocytes was increased. Optic nerve from EAE and optic chiasm from MS also showed decreased cholesterol synthesis gene expression. The potential role of cholesterol synthesized by astrocytes during EAE and MS is discussed. Together, this provides proof-of-concept that a cell-specific and region-specific gene expression approach can provide potential treatment targets in distinct neuroanatomic regions during multifocal neurological diseases.
Collapse
|
793
|
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: 156] [Impact Index Per Article: 22.3] [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
|
794
|
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: 61] [Impact Index Per Article: 8.7] [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
|
795
|
Progressive striatonigral degeneration in a transgenic mouse model of multiple system atrophy: translational implications for interventional therapies. Acta Neuropathol Commun 2018; 6:2. [PMID: 29298733 PMCID: PMC5753576 DOI: 10.1186/s40478-017-0504-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/16/2017] [Indexed: 12/31/2022] Open
Abstract
Multiple system atrophy (MSA) is a rapidly progressive neurodegenerative disorder characterized by widespread oligodendroglial cytoplasmic inclusions of filamentous α-synuclein, and neuronal loss in autonomic centres, basal ganglia and cerebellar circuits. It has been suggested that primary oligodendroglial α-synucleinopathy may represent a trigger in the pathogenesis of MSA, but the mechanisms underlying selective vulnerability and disease progression are unclear. The post-mortem analysis of MSA brains provides a static final picture of the disease neuropathology, but gives no clear indication on the sequence of pathogenic events in MSA. Therefore, alternative methods are needed to address these issues. We investigated selective vulnerability and disease progression in the transgenic PLP-α-syn mouse model of MSA characterized by targeted oligodendroglial α-synuclein overexpression aiming to provide a neuropathological correlate of motor deterioration. We show progressive motor deficits that emerge at 6 months of age and deteriorate up to 18 months of follow-up. The motor phenotype was associated with dopaminergic cell loss in the substantia nigra pars compacta at 6 months, followed by loss of striatal dopaminergic terminals and DARPP32-positive medium sized projection neurons at 12 months. Olivopontocerebellar motor loops remained spared in the PLP-α-syn model of MSA. These findings replicate progressive striatonigral degeneration underlying Parkinson-variant MSA. The initiation of the degenerative process was linked to an increase of soluble oligomeric α-synuclein species between 2 and 6 months. Early region-specific α-synuclein-associated activation profile of microglia was found in MSA substantia nigra. The role of abnormal neuroinflammatory signalling in disease progression was further supported by increased levels of CD68, CCL3, CCL5 and M-CSF with a peak in aged PLP-α-syn mice. In summary, transgenic PLP-α-syn mice show a distinctive oligodendroglial α-synucleinopathy that is associated with progressive striatonigral degeneration linked to abnormal neuroinflammatory response. The model provides a relevant tool for preclinical therapeutic target discovery for human Parkinson-variant MSA.
Collapse
|
796
|
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
|
797
|
Maduna T, Audouard E, Dembélé D, Mouzaoui N, Reiss D, Massotte D, Gaveriaux-Ruff C. Microglia Express Mu Opioid Receptor: Insights From Transcriptomics and Fluorescent Reporter Mice. Front Psychiatry 2018; 9:726. [PMID: 30662412 PMCID: PMC6328486 DOI: 10.3389/fpsyt.2018.00726] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Microglia activation contributes to chronic pain and to the adverse effects of opiate use such as analgesic tolerance and opioid-induced hyperalgesia. Both mu opioid receptor (MOR) encoded by Oprm1/OPRM1 gene and toll like receptor 4 (TLR4) have been reported to mediate these morphine effects and a current question is whether microglia express the Oprm1 transcript and MOR protein. The aim of this study was to characterize Oprm1-MOR expression in naive murine and human microglia, combining transcriptomics datasets previously published by other groups with our own imaging study using the Cx3cr1-eGFP-MOR-mCherry reporter mouse line. Methods: We analyzed microglial Oprm1/OPRM1 expression obtained from transcriptomics datasets, focusing on ex vivo studies from adult wild-type animals and adult post-mortem human cerebral cortex. Oprm1, as well as co-regulated gene sets were examined. The expression of MOR in microglia was also investigated using our novel fluorescent Cx3cr1-eGFP-MOR-mcherry reporter mouse line. We determined whether CX3cR1-eGFP positive microglial cells expressed MOR-mCherry protein by imaging various brain areas including the Frontal Cortex, Nucleus Accumbens, Ventral Tegmental Area, Central Amygdala, and Periaqueductal Gray matter, as well as spinal cord. Results: Oprm1 expression was found in all 12 microglia datasets from mouse whole brain, in 7 out of 8 from cerebral cortex, 3 out of 4 from hippocampus, 1 out of 1 from striatum, and 4 out of 5 from mouse or rat spinal cord. OPRM1 was expressed in 16 out of 17 microglia transcriptomes from human cerebral cortex. In Cx3cr1-eGFP-MOR-mCherry mice, the percentage of MOR-positive microglial cells ranged between 35.4 and 51.6% in the different brain areas, and between 36.8 and 42.4% in the spinal cord. Conclusion: The comparative analysis of the microglia transcriptomes indicates that Oprm1/OPRM1 transcripts are expressed in microglia. The investigation of Cx3cr1-eGFP-MOR-mCherry mice also shows microglial expression of MOR proteinin the brain and spine. These results corroborate functional studies showing the actions of MOR agonists on microglia and suppression of these effects by MOR-selective antagonists or MOR knockdown.
Collapse
Affiliation(s)
- Tando Maduna
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Emilie Audouard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Doulaye Dembélé
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Nejma Mouzaoui
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - David Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Dominique Massotte
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Université de Strasbourg, Strasbourg, France
| | - Claire Gaveriaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| |
Collapse
|
798
|
Koso H, Nishinakamura R, Watanabe S. Sall1 Regulates Microglial Morphology Cell Autonomously in the Developing Retina. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1074:209-215. [PMID: 29721946 DOI: 10.1007/978-3-319-75402-4_26] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Retinal degeneration often accompanies microglial activation and infiltration of monocyte-derived macrophages into the retina, resulting in the coexistence of microglia and monocyte-derived macrophages in the retina. We previously showed that the Sall1 zinc-finger transcriptional factor is expressed specifically in microglia within the retinal phagocyte pool, and analyses of Sall1 knockout mice revealed that microglial morphology changed from a ramified to a more amoeboid appearance in the developing retina. To investigate further whether Sall1 functions autonomously in microglia, we generated Sall1 conditional knockout mice, in which Sall1 was depleted specifically in the Cx3cr1+ microglial compartment of the developing retina. Sall1-deficient microglia exhibited morphological abnormalities on embryonic day 18 that strikingly resembled the phenotype observed in Sall1 knockout mice, demonstrating that Sall1 regulates microglial morphology cell autonomously. Analysis of the postnatal retina revealed that Sall1-deficient microglia extended their processes and their morphology became comparable to that of wild-type microglia on postnatal day 21, indicating that Sall1 is essential for microglial ramification in the developing retina, but not in the postnatal retina.
Collapse
Affiliation(s)
- Hideto Koso
- Division of Molecular and Developmental Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Sumiko Watanabe
- Division of Molecular and Developmental Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
799
|
Kurotaki D, Sasaki H, Tamura T. Transcriptional control of monocyte and macrophage development. Int Immunol 2018; 29:97-107. [PMID: 28379391 DOI: 10.1093/intimm/dxx016] [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: 03/13/2017] [Accepted: 03/19/2017] [Indexed: 12/12/2022] Open
Abstract
Monocytes and macrophages play critical roles in immune responses, tissue homeostasis and disease progression. There are a number of functionally and phenotypically distinct subpopulations throughout the body. However, the mechanisms by which macrophage and monocyte heterogeneity is established remain unclear. Recent studies have suggested that most tissue-resident macrophages originate from fetal progenitors but not from hematopoietic stem cells, whereas some subpopulations are derived from adult monocytes. In addition, transcription factors specifically required for the development of each subpopulation have been identified. Interestingly, local environmental factors such as heme, retinoic acid and RANKL induce the expression and/or activation of tissue-specific transcription factors, thereby controlling transcriptional programs specific for the subpopulations. Thus, distinct differentiation pathways and local microenvironments appear to contribute to the determination of macrophage transcriptional identities. In this review, we highlight recent advances in our knowledge of the transcriptional control of macrophage and monocyte development.
Collapse
Affiliation(s)
- Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Haruka Sasaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| |
Collapse
|
800
|
Mizee MR, Poel MVD, Huitinga I. Purification of cells from fresh human brain tissue: primary human glial cells. HANDBOOK OF CLINICAL NEUROLOGY 2018; 150:273-283. [PMID: 29496146 DOI: 10.1016/b978-0-444-63639-3.00019-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In order to translate the findings obtained from postmortem brain tissue samples to functional biologic mechanisms of central nervous system disease, it will be necessary to understand how these findings affect the different cell populations in the brain. The acute isolation and analysis of pure glial cell populations are common practice in animal models for neurologic diseases, but are not yet regularly applied to human postmortem brain material. The development of novel cell isolation techniques and methods for transcriptomic and proteomic analysis have made it possible to isolate and phenotype primary human cell populations from the central nervous system. The psychiatric program of the Netherlands Brain Bank has considerable experience with the purification of glial cells. This chapter will review the rapid isolation and phenotyping procedures for two major glia cell populations in the human brain, microglia and astrocytes, and will also discuss the potential for biobanking these cells, as well as the possible alternatives to cell isolations. The acute isolation of glial cells without culture-based adherence steps allows the analysis of glial alterations that underlie, or are the result of, disease neuropathology of the donor.
Collapse
Affiliation(s)
- Mark Ronald Mizee
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; Psychiatric Program, Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Marlijn van der Poel
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Inge Huitinga
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; Psychiatric Program, Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|