201
|
Wu Q, Shi J, Ji X, Xia T, Zeng L, Li G, Wang Y, Gao J, Yao L, Ma J, Liu X, Liu N, Hu L, He B, Liang Y, Qu G, Jiang G. Heterogenous Internalization of Nanoparticles at Ultra-Trace Concentration in Environmental Individual Unicellular Organisms Unveiled by Single-Cell Mass Cytometry. ACS NANO 2020; 14:12828-12839. [PMID: 32931264 DOI: 10.1021/acsnano.0c03587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The application and consumption of nanoparticles (NPs) inevitably result in the contamination of environmental water. The internalized NPs in unicellular organisms could travel to human bodies along food chains and raise health concerns. Current research failed to determine the characteristics of cellular uptake of NPs by unicellular organisms at extremely low concentration in the real environment. We here developed a label-free high-throughput mass cytometry method to investigate gold NP (AuNP) uptake in a unicellular organism (Tetrahymena thermophila) at the single-cell level. The limit of detection for Au is as low as to 6.67 × 10-18 g/cell, which equals ∼5.3 5 nm AuNPs. We demonstrated that active engulfment pathways were responsible for the cellular accumulation of AuNPs and T. thermophila could also eliminate the cellular AuNPs rapidly. The interaction between AuNPs and T. thermophila is highly dependent on the sizes of nanoparticles; i.e., the population of T. thermophila containing AuNPs decreased with the increment of the diameters of AuNPs when exposed to the same mass concentration. For each type of AuNP, distinct heterogeneous cellular uptake of AuNPs by T. thermophila was observed. Intriguingly, for 5 nm AuNP, even at 0.001 ng/mL, some T. thermophila cells could concentrate AuNPs, indicating a real environmental concern even when water was contaminated by only trace level of NPs. This method represents a promising tool for simultaneous determination of physiological status of cells together with the intracellular level of heavy metal or metallic NPs in study of biological effects.
Collapse
Affiliation(s)
- Qi Wu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Jianbo Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Xiaomeng Ji
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Li Zeng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Gengtan Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Yuanyuan Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Jie Gao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Linlin Yao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Junjie Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Xiaolei Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Nian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Ligang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Bin He
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Yong Liang
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
- University of Chinese Academy of Science, Beijing, 100049, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| |
Collapse
|
202
|
Enzymatic Dissociation Induces Transcriptional and Proteotype Bias in Brain Cell Populations. Int J Mol Sci 2020; 21:ijms21217944. [PMID: 33114694 PMCID: PMC7663484 DOI: 10.3390/ijms21217944] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022] Open
Abstract
Different cell isolation techniques exist for transcriptomic and proteotype profiling of brain cells. Here, we provide a systematic investigation of the influence of different cell isolation protocols on transcriptional and proteotype profiles in mouse brain tissue by taking into account single-cell transcriptomics of brain cells, proteotypes of microglia and astrocytes, and flow cytometric analysis of microglia. We show that standard enzymatic digestion of brain tissue at 37 °C induces profound and consistent alterations in the transcriptome and proteotype of neuronal and glial cells, as compared to an optimized mechanical dissociation protocol at 4 °C. These findings emphasize the risk of introducing technical biases and biological artifacts when implementing enzymatic digestion-based isolation methods for brain cell analyses.
Collapse
|
203
|
Cipollina G, Davari Serej A, Di Nolfi G, Gazzano A, Marsala A, Spatafora MG, Peviani M. Heterogeneity of Neuroinflammatory Responses in Amyotrophic Lateral Sclerosis: A Challenge or an Opportunity? Int J Mol Sci 2020; 21:E7923. [PMID: 33113845 PMCID: PMC7662281 DOI: 10.3390/ijms21217923] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex pathology: (i) the neurodegeneration is chronic and progressive; it starts focally in specific central nervous system (CNS) areas and spreads to different districts; (ii) multiple cell types further than motor neurons (i.e., glial/immune system cells) are actively involved in the disease; (iii) both neurosupportive and neurotoxic neuroinflammatory responses were identified. Microglia cells (a key player of neuroinflammation in the CNS) attracted great interest as potential target cell population that could be modulated to counteract disease progression, at least in preclinical ALS models. However, the heterogeneous/multifaceted microglia cell responses occurring in different CNS districts during the disease represent a hurdle for clinical translation of single-drug therapies. To address this issue, over the past ten years, several studies attempted to dissect the complexity of microglia responses in ALS. In this review, we shall summarize these results highlighting how the heterogeneous signature displayed by ALS microglia reflects not only the extent of neuronal demise in different regions of the CNS, but also variable engagement in the attempts to cope with the neuronal damage. We shall discuss novel avenues opened by the advent of single-cell and spatial transcriptomics technologies, underlining the potential for discovery of novel therapeutic targets, as well as more specific diagnostic/prognostic not-invasive markers of neuroinflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marco Peviani
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, via Ferrata 9, 27100 Pavia, Italy; (G.C.); (A.D.S.); (G.D.N.); (A.G.); (A.M.); (M.G.S.)
| |
Collapse
|
204
|
Miedema A, Wijering MHC, Eggen BJL, Kooistra SM. High-Resolution Transcriptomic and Proteomic Profiling of Heterogeneity of Brain-Derived Microglia in Multiple Sclerosis. Front Mol Neurosci 2020; 13:583811. [PMID: 33192299 PMCID: PMC7654237 DOI: 10.3389/fnmol.2020.583811] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Microglia are important for central nervous system (CNS) homeostasis and first to respond to tissue damage and perturbations. Microglia are heterogeneous cells; in case of pathology, microglia adopt a range of phenotypes with altered functions. However, how these different microglia subtypes are implicated in CNS disease is largely unresolved. Multiple sclerosis (MS) is a chronic demyelinating disease of the CNS, characterized by inflammation and axonal degeneration, ultimately leading to neurological decline. One way microglia are implicated in MS is through stimulation of remyelination. They facilitate efficient remyelination by phagocytosis of myelin debris. In addition, microglia recruit oligodendrocyte precursor cells (OPCs) to demyelinated areas and stimulate remyelination. The development of high-resolution technologies to profile individual cells has greatly contributed to our understanding of microglia heterogeneity and function under normal and pathological conditions. Gene expression profiling technologies have evolved from whole tissue RNA sequencing toward single-cell or nucleus sequencing. Single microglia proteomic profiles are also increasingly generated, offering another layer of high-resolution data. Here, we will review recent studies that have employed these technologies in the context of MS and their respective advantages and disadvantages. Moreover, recent developments that allow for (single) cell profiling while retaining spatial information and tissue context will be discussed.
Collapse
Affiliation(s)
- Anneke Miedema
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marion H C Wijering
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bart J L Eggen
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Susanne M Kooistra
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
205
|
Banerjee P, Paza E, Perkins EM, James OG, Kenkhuis B, Lloyd AF, Burr K, Story D, Yusuf D, He X, Backofen R, Dando O, Chandran S, Priller J. Generation of pure monocultures of human microglia-like cells from induced pluripotent stem cells. Stem Cell Res 2020; 49:102046. [PMID: 33096385 DOI: 10.1016/j.scr.2020.102046] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 01/28/2023] Open
Abstract
Microglia are resident tissue macrophages of the central nervous system (CNS) that arise from erythromyeloid progenitors during embryonic development. They play essential roles in CNS development, homeostasis and response to disease. Since microglia are difficult to procure from the human brain, several protocols have been developed to generate microglia-like cells from human induced pluripotent stem cells (hiPSCs). However, some concerns remain over the purity and quality of in vitro generated microglia. Here, we describe a new protocol that does not require co-culture with neural cells and yields cultures of 100% P2Y12+ 95% TMEM119+ ramified human microglia-like cells (hiPSC-MG). In the presence of neural precursor cell-conditioned media, hiPSC-MG expressed high levels of human microglia signature genes, including SALL1, CSF1R, P2RY12, TMEM119, TREM2, HEXB and SIGLEC11, as revealed by whole-transcriptome analysis. Stimulation of hiPSC-MG with lipopolysaccharide resulted in downregulation of P2Y12 expression, induction of IL1B mRNA expression and increase in cell capacitance. HiPSC-MG were phagocytically active and maintained their cell identity after transplantation into murine brain slices and human brain spheroids. Together, our new protocol for the generation of microglia-like cells from human iPSCs will facilitate the study of human microglial function in health and disease.
Collapse
Affiliation(s)
- Poulomi Banerjee
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Evdokia Paza
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Emma M Perkins
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Owen G James
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Boyd Kenkhuis
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Amy F Lloyd
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Karen Burr
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - David Story
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Dilmurat Yusuf
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Freiburg, Germany
| | - Xin He
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Rolf Backofen
- Bioinformatics Group, Department of Computer Science, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Owen Dando
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Josef Priller
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK; Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité, Universitätsmedizin Berlin, BIH and DZNE, Berlin, Germany.
| |
Collapse
|
206
|
Perea JR, Bolós M, Avila J. Microglia in Alzheimer's Disease in the Context of Tau Pathology. Biomolecules 2020; 10:biom10101439. [PMID: 33066368 PMCID: PMC7602223 DOI: 10.3390/biom10101439] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022] Open
Abstract
Microglia are the cells that comprise the innate immune system in the brain. First described more than a century ago, these cells were initially assigned a secondary role in the central nervous system (CNS) with respect to the protagonists, neurons. However, the latest advances have revealed the complexity and importance of microglia in neurodegenerative conditions such as Alzheimer’s disease (AD), the most common form of dementia associated with aging. This pathology is characterized by the accumulation of amyloid-β peptide (Aβ), which forms senile plaques in the neocortex, as well as by the aggregation of hyperphosphorylated tau protein, a process that leads to the development of neurofibrillary tangles (NFTs). Over the past few years, efforts have been focused on studying the interaction between Aβ and microglia, together with the ability of the latter to decrease the levels of this peptide. Given that most clinical trials following this strategy have failed, current endeavors focus on deciphering the molecular mechanisms that trigger the tau-induced inflammatory response of microglia. In this review, we summarize the most recent studies on the physiological and pathological functions of tau protein and microglia. In addition, we analyze the impact of microglial AD-risk genes (APOE, TREM2, and CD33) in tau pathology, and we discuss the role of extracellular soluble tau in neuroinflammation.
Collapse
Affiliation(s)
- Juan Ramón Perea
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 1 Nicolás Cabrera, 28049 Madrid, Spain; (J.R.P.); (M.B.)
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), 5 Valderrebollo, 28031 Madrid, Spain
| | - Marta Bolós
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 1 Nicolás Cabrera, 28049 Madrid, Spain; (J.R.P.); (M.B.)
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), 5 Valderrebollo, 28031 Madrid, Spain
| | - Jesús Avila
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 1 Nicolás Cabrera, 28049 Madrid, Spain; (J.R.P.); (M.B.)
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), 5 Valderrebollo, 28031 Madrid, Spain
- Correspondence: ; Tel.:+34-196-4564
| |
Collapse
|
207
|
Frank S. Catch me if you can: SARS-CoV-2 detection in brains of deceased patients with COVID-19. Lancet Neurol 2020; 19:883-884. [PMID: 33031734 PMCID: PMC7535625 DOI: 10.1016/s1474-4422(20)30371-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Stephan Frank
- Division of Neuropathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland.
| |
Collapse
|
208
|
Zabegalov KN, Wang D, Yang L, Wang J, Hu G, Serikuly N, Alpyshov ET, Khatsko SL, Zhdanov A, Demin KA, Galstyan DS, Volgin AD, de Abreu MS, Strekalova T, Song C, Amstislavskaya TG, Sysoev Y, Musienko PE, Kalueff AV. Decoding the role of zebrafish neuroglia in CNS disease modeling. Brain Res Bull 2020; 166:44-53. [PMID: 33027679 DOI: 10.1016/j.brainresbull.2020.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/14/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
Abstract
Neuroglia, including microglia and astrocytes, is a critical component of the central nervous system (CNS) that interacts with neurons to modulate brain activity, development, metabolism and signaling pathways. Thus, a better understanding of the role of neuroglia in the brain is critical. Complementing clinical and rodent data, the zebrafish (Danio rerio) is rapidly becoming an important model organism to probe the role of neuroglia in brain disorders. With high genetic and physiological similarity to humans and rodents, zebrafish possess some common (shared), as well as some specific molecular biomarkers and features of neuroglia development and functioning. Studying these common and zebrafish-specific aspects of neuroglia may generate important insights into key brain mechanisms, including neurodevelopmental, neurodegenerative, neuroregenerative and neurological processes. Here, we discuss the biology of neuroglia in humans, rodents and fish, its role in various CNS functions, and further directions of translational research into the role of neuroglia in CNS disorders using zebrafish models.
Collapse
Affiliation(s)
- Konstantin N Zabegalov
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia
| | - Dongmei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - LongEn Yang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Jingtao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Guojun Hu
- School of Pharmacy, Southwest University, Chongqing, China
| | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | | | | | | | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia.
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Division of Molecular Psychiatry, Centre of Mental Health, University of Würzburg, Würzburg, Germany
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Tamara G Amstislavskaya
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Zelman Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Yury Sysoev
- Laboratory of Neuroprosthetics, Institute of Translational Biomedicine, Petersburg State University, St. Petersburg, Russia; Department of Pharmacology and Clinical Pharmacology, St. Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia
| | - Pavel E Musienko
- Laboratory of Neuroprosthetics, Institute of Translational Biomedicine, Petersburg State University, St. Petersburg, Russia; Institute of Phthisiopulmonology, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
209
|
Zhang T, Warden AR, Li Y, Ding X. Progress and applications of mass cytometry in sketching immune landscapes. Clin Transl Med 2020; 10:e206. [PMID: 33135337 PMCID: PMC7556381 DOI: 10.1002/ctm2.206] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Recently emerged mass cytometry (cytometry by time-of-flight [CyTOF]) technology permits the identification and quantification of inherently diverse cellular systems, and the simultaneous measurement of functional attributes at the single-cell resolution. By virtue of its multiplex ability with limited need for compensation, CyTOF has led a critical role in immunological research fields. Here, we present an overview of CyTOF, including the introduction of CyTOF principle and advantages that make it a standalone tool in deciphering immune mysteries. We then discuss the functional assays, introduce the bioinformatics to interpret the data yield via CyTOF, and depict the emerging clinical and research applications of CyTOF technology in sketching immune landscape in a wide variety of diseases.
Collapse
Affiliation(s)
- Ting Zhang
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Antony R. Warden
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Yiyang Li
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Xianting Ding
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
210
|
Provenzano F, Pérez MJ, Deleidi M. Redefining Microglial Identity in Health and Disease at Single-Cell Resolution. Trends Mol Med 2020; 27:47-59. [PMID: 33008729 DOI: 10.1016/j.molmed.2020.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022]
Abstract
Microglia have long been considered a homogenous cell population that uniformly responds to extrinsic factors. Here, we describe how the recent development of single-cell technologies has revealed the heterogeneity of both human and mouse microglia and identified distinct microglial states linked to specific developmental, aging, and disease stages. We discuss progress and future developments in data analysis, essential tools for the comprehension of big data derived from single-cell omics, and the necessity of integrating such data with functional studies to correlate genetic cues with the relevant biological functions of microglia. Defining the functional correlates of distinct microglia states is fundamental to dissecting the 'microglial etiology' of aging and complex neurological diseases and identifying novel therapeutic and diagnostic targets.
Collapse
Affiliation(s)
- Francesca Provenzano
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - María José Pérez
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Michela Deleidi
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
211
|
Single-cell mass cytometry of microglia in major depressive disorder reveals a non-inflammatory phenotype with increased homeostatic marker expression. Transl Psychiatry 2020; 10:310. [PMID: 32917850 PMCID: PMC7486938 DOI: 10.1038/s41398-020-00992-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/10/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
Stress-induced disturbances of brain homeostasis and neuroinflammation have been implicated in the pathophysiology of mood disorders. In major depressive disorder (MDD), elevated levels of proinflammatory cytokines and chemokines can be found in peripheral blood, but very little is known about the changes that occur directly in the brain. Microglia are the primary immune effector cells of the central nervous system and exquisitely sensitive to changes in the brain microenvironment. Here, we performed the first single-cell analysis of microglia from four different post-mortem brain regions (frontal lobe, temporal lobe, thalamus, and subventricular zone) of medicated individuals with MDD compared to controls. We found no evidence for the induction of inflammation-associated molecules, such as CD11b, CD45, CCL2, IL-1β, IL-6, TNF, MIP-1β (CCL4), IL-10, and even decreased expression of HLA-DR and CD68 in microglia from MDD cases. In contrast, we detected increased levels of the homeostatic proteins P2Y12 receptor, TMEM119 and CCR5 (CD195) in microglia from all brain regions of individuals with MDD. We also identified enrichment of non-inflammatory CD206hi macrophages in the brains of MDD cases. In sum, our results suggest enhanced homeostatic functions of microglia in MDD.
Collapse
|
212
|
Bagdas D, Paris JJ, Carper M, Wodarski R, Rice AS, Knapp PE, Hauser KF, Damaj MI. Conditional expression of HIV-1 tat in the mouse alters the onset and progression of tonic, inflammatory and neuropathic hypersensitivity in a sex-dependent manner. Eur J Pain 2020; 24:1609-1623. [PMID: 32533878 PMCID: PMC7856573 DOI: 10.1002/ejp.1618] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 05/21/2020] [Accepted: 06/05/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND At least one-third of HIV-1-afflicted individuals experience peripheral neuropathy. Although the underlying mechanisms are not known, they may involve neurotoxic HIV-1 proteins. METHODS We assessed the influence of the neurotoxic HIV-1 regulatory protein, Tat, on inflammatory and neuropathic nociceptive behaviours using transgenic male and female mice that conditionally expressed (or did not express) HIV-1 Tat1-86 in fibrillary acidic protein-expressing glia in the central and peripheral nervous systems. RESULTS Tat induction significantly attenuated the time spent paw-licking following formalin injection (2.5%, i.pl.) in both male and female mice. However, significant sex differences were observed in the onset and magnitude of inflammation and sensory sensitivity following complete Freund's adjuvant (CFA) injection (10%, i.pl.) after Tat activation. Unlike female mice, male mice showed a significant attenuation of paw swelling and an absence of mechanical/thermal hypersensitivity in response to CFA after Tat induction. Male Tat(+) mice also showed accelerated recovery from chronic constrictive nerve injury (CCI)-induced neuropathic mechanical and thermal hypersensitivity compared to female Tat(+) mice. Morphine (3.2 mg/kg) fully reversed CCI-induced mechanical hypersensitivity in female Tat(-) mice, but not in Tat(+) females. CONCLUSIONS The ability of Tat to decrease oedema, paw swelling, and limit allodynia suggests a sequel of events in which Tat-induced functional deficits precede the onset of mechanical hypersensitivity. Moreover, HIV-1 Tat attenuated responses to inflammatory and neuropathic insults in a sex-dependent manner. HIV-1 Tat appears to directly contribute to HIV sensory neuropathy and reveals sex differences in HIV responsiveness and/or the underlying peripheral neuroinflammatory and nociceptive mechanisms.
Collapse
Affiliation(s)
- Deniz Bagdas
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- The Center for the Study for Tobacco Products, Virginia Commonwealth University, Richmond, VA 23284-2018, USA
| | - Jason J. Paris
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA
- Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA
| | - Moriah Carper
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Rachel Wodarski
- Pain Research Group, Department of Surgery & Cancer, Imperial College, London, SW10 9NH, UK
| | - Andrew S.C. Rice
- Pain Research Group, Department of Surgery & Cancer, Imperial College, London, SW10 9NH, UK
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Department of Anatomy & Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Institute for Drug and Alcohol Studies, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Department of Anatomy & Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- Institute for Drug and Alcohol Studies, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - M. Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
- The Center for the Study for Tobacco Products, Virginia Commonwealth University, Richmond, VA 23284-2018, USA
- Translational Research Initiative for Pain and Neuropathy at VCU, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| |
Collapse
|
213
|
Böttcher C, van der Poel M, Fernández-Zapata C, Schlickeiser S, Leman JKH, Hsiao CC, Mizee MR, Adelia, Vincenten MCJ, Kunkel D, Huitinga I, Hamann J, Priller J. Single-cell mass cytometry reveals complex myeloid cell composition in active lesions of progressive multiple sclerosis. Acta Neuropathol Commun 2020; 8:136. [PMID: 32811567 PMCID: PMC7437178 DOI: 10.1186/s40478-020-01010-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/01/2020] [Indexed: 01/15/2023] Open
Abstract
Myeloid cells contribute to inflammation and demyelination in the early stages of multiple sclerosis (MS), but it is still unclear to what extent these cells are involved in active lesion formation in progressive MS (PMS). Here, we have harnessed the power of single-cell mass cytometry (CyTOF) to compare myeloid cell phenotypes in active lesions of PMS donors with those in normal-appearing white matter from the same donors and control white matter from non-MS donors. CyTOF measurements of a total of 74 targeted proteins revealed a decreased abundance of homeostatic and TNFhi microglia, and an increase in highly phagocytic and activated microglia states in active lesions of PMS donors. Interestingly, in contrast to results obtained from studies of the inflammatory early disease stages of MS, infiltrating monocyte-derived macrophages were scarce in active lesions of PMS, suggesting fundamental differences of myeloid cell composition in advanced stages of PMS.
Collapse
Affiliation(s)
- Chotima Böttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Marlijn van der Poel
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Camila Fernández-Zapata
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stephan Schlickeiser
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia K H Leman
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cheng-Chih Hsiao
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark R Mizee
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Adelia
- Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Maria C J Vincenten
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Desiree Kunkel
- Flow & Mass Cytometry Core Facility, Charité - Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Inge Huitinga
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Jörg Hamann
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.
- University of Edinburgh and UK Dementia Research Institute (DRI), Edinburgh, UK.
| |
Collapse
|
214
|
Hanger B, Couch A, Rajendran L, Srivastava DP, Vernon AC. Emerging Developments in Human Induced Pluripotent Stem Cell-Derived Microglia: Implications for Modelling Psychiatric Disorders With a Neurodevelopmental Origin. Front Psychiatry 2020; 11:789. [PMID: 32848951 PMCID: PMC7433763 DOI: 10.3389/fpsyt.2020.00789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/23/2020] [Indexed: 12/15/2022] Open
Abstract
Microglia, the resident tissue macrophages of the brain, are increasingly implicated in the pathophysiology of psychiatric disorders with a neurodevelopmental origin, including schizophrenia. To date, however, our understanding of the potential role for these cells in schizophrenia has been informed by studies of aged post-mortem samples, low resolution in vivo neuroimaging and rodent models. Whilst these have provided important insights, including signs of the heterogeneous nature of microglia, we currently lack a validated human in vitro system to characterize microglia in the context of brain health and disease during neurodevelopment. Primarily, this reflects a lack of access to human primary tissue during developmental stages. In this review, we first describe microglia, including their ontogeny and heterogeneity and consider their role in brain development. We then provide an evaluation of the potential for differentiating microglia from human induced pluripotent stem cells (hiPSCs) as a robust in vitro human model system to study these cells. We find the majority of protocols for hiPSC-derived microglia generate cells characteristically similar to foetal stage microglia when exposed to neuronal environment-like cues. This may represent a robust and relevant model for the study of cellular and molecular mechanisms in schizophrenia. Each protocol however, provides unique benefits as well as shortcomings, highlighting the need for context-dependent protocol choice and cross-lab collaboration and communication to identify the most robust and translatable microglia model.
Collapse
Affiliation(s)
- Bjørn Hanger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Amalie Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Lawrence Rajendran
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Anthony C. Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| |
Collapse
|
215
|
Jurga AM, Paleczna M, Kuter KZ. Overview of General and Discriminating Markers of Differential Microglia Phenotypes. Front Cell Neurosci 2020; 14:198. [PMID: 32848611 PMCID: PMC7424058 DOI: 10.3389/fncel.2020.00198] [Citation(s) in RCA: 569] [Impact Index Per Article: 113.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammatory processes and microglia activation accompany most of the pathophysiological diseases in the central nervous system. It is proven that glial pathology precedes and even drives the development of multiple neurodegenerative conditions. A growing number of studies point out the importance of microglia in brain development as well as in physiological functioning. These resident brain immune cells are divergent from the peripherally infiltrated macrophages, but their precise in situ discrimination is surprisingly difficult. Microglial heterogeneity in the brain is especially visible in their morphology and cell density in particular brain structures but also in the expression of cellular markers. This often determines their role in physiology or pathology of brain functioning. The species differences between rodent and human markers add complexity to the whole picture. Furthermore, due to activation, microglia show a broad spectrum of phenotypes ranging from the pro-inflammatory, potentially cytotoxic M1 to the anti-inflammatory, scavenging, and regenerative M2. A precise distinction of specific phenotypes is nowadays essential to study microglial functions and tissue state in such a quickly changing environment. Due to the overwhelming amount of data on multiple sets of markers that is available for such studies, the choice of appropriate markers is a scientific challenge. This review gathers, classifies, and describes known and recently discovered protein markers expressed by microglial cells in their different phenotypes. The presented microglia markers include qualitative and semi-quantitative, general and specific, surface and intracellular proteins, as well as secreted molecules. The information provided here creates a comprehensive and practical guide through the current knowledge and will facilitate the choosing of proper, more specific markers for detailed studies on microglia and neuroinflammatory mechanisms in various physiological as well as pathological conditions. Both basic research and clinical medicine need clearly described and validated molecular markers of microglia phenotype, which are essential in diagnostics, treatment, and prevention of diseases engaging glia activation.
Collapse
Affiliation(s)
- Agnieszka M Jurga
- Maj Institute of Pharmacology, Department of Neuropsychopharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Martyna Paleczna
- Maj Institute of Pharmacology, Department of Neuropsychopharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Z Kuter
- Maj Institute of Pharmacology, Department of Neuropsychopharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
216
|
Jäkel S, Williams A. What Have Advances in Transcriptomic Technologies Taught us About Human White Matter Pathologies? Front Cell Neurosci 2020; 14:238. [PMID: 32848627 PMCID: PMC7418269 DOI: 10.3389/fncel.2020.00238] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/07/2020] [Indexed: 11/16/2022] Open
Abstract
For a long time, post-mortem analysis of human brain pathologies has been purely descriptive, limiting insight into the pathological mechanisms. However, starting in the early 2000s, next-generation sequencing (NGS) and the routine application of bulk RNA-sequencing and microarray technologies have revolutionized the usefulness of post-mortem human brain tissue. This has allowed many studies to provide novel mechanistic insights into certain brain pathologies, albeit at a still unsatisfying resolution, with masking of lowly expressed genes and regulatory elements in different cell types. The recent rapid evolution of single-cell technologies has now allowed researchers to shed light on human pathologies at a previously unreached resolution revealing further insights into pathological mechanisms that will open the way for the development of new strategies for therapies. In this review article, we will give an overview of the incremental information that single-cell technologies have given us for human white matter (WM) pathologies, summarize which single-cell technologies are available, and speculate where these novel approaches may lead us for pathological assessment in the future.
Collapse
Affiliation(s)
- Sarah Jäkel
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | | |
Collapse
|
217
|
The Role of TGFβ Signaling in Microglia Maturation and Activation. Trends Immunol 2020; 41:836-848. [PMID: 32741652 DOI: 10.1016/j.it.2020.07.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/23/2022]
Abstract
The pleiotropic cytokine transforming growth factor-beta 1 (TGFβ1) plays pivotal roles in different cell types, including immune cells such as T cells, monocytes/macrophages, and microglia. Microglia are essential during physiological and pathological events. Maturation of postnatal microglia, as well as the regulation of the complex functional repertoire of microglia, needs to be carefully orchestrated. However, an understanding of how mammalian microglia maturation and disease-associated microglia activation is regulated remains fragmentary. Here, we summarize recent observations made by employing transgenic approaches to silence microglial TGFβ signaling in mice. These revealed that TGFβ1 and TGFβ signaling are indispensable for microglia maturation, adult microglia homeostasis, and the control of microglia activation in central nervous system pathologies.
Collapse
|
218
|
Miron VE, Priller J. Investigating Microglia in Health and Disease: Challenges and Opportunities. Trends Immunol 2020; 41:785-793. [PMID: 32736967 DOI: 10.1016/j.it.2020.07.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/24/2022]
Abstract
Microglia are tissue-resident macrophages implicated in central nervous system (CNS) development, homeostasis, and response to injury. Recent advances in transcriptomics, multiplex protein expression analysis, and experimental depletion of microglia have cemented their importance. However, it is still unclear which models are best suited to investigate microglia and explore their function in human disease. Here, we discuss issues regarding off-targeting during experimental manipulation, and differences and similarities between human and rodent microglia. With new developments in transgenic lines and human-rodent chimeras, we anticipate that in coming years, a clearer picture of microglia function in health and disease will emerge.
Collapse
Affiliation(s)
- Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK.
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin and DZNE, Berlin, Germany; UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
219
|
Althammer F, Ferreira-Neto HC, Rubaharan M, Roy RK, Patel AA, Murphy A, Cox DN, Stern JE. Three-dimensional morphometric analysis reveals time-dependent structural changes in microglia and astrocytes in the central amygdala and hypothalamic paraventricular nucleus of heart failure rats. J Neuroinflammation 2020; 17:221. [PMID: 32703230 PMCID: PMC7379770 DOI: 10.1186/s12974-020-01892-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cardiovascular diseases, including heart failure, are the most common cause of death globally. Recent studies support a high degree of comorbidity between heart failure and cognitive and mood disorders resulting in memory loss, depression, and anxiety. While neuroinflammation in the hypothalamic paraventricular nucleus contributes to autonomic and cardiovascular dysregulation in heart failure, mechanisms underlying cognitive and mood disorders in this disease remain elusive. The goal of this study was to quantitatively assess markers of neuroinflammation (glial morphology, cytokines, and A1 astrocyte markers) in the central amygdala, a critical forebrain region involved in emotion and cognition, and to determine its time course and correlation to disease severity during the progression of heart failure. METHODS We developed and implemented a comprehensive microglial/astrocyte profiler for precise three-dimensional morphometric analysis of individual microglia and astrocytes in specific brain nuclei at different time points during the progression of heart failure. To this end, we used a well-established ischemic heart failure rat model. Morphometric studies were complemented with quantification of various pro-inflammatory cytokines and A1/A2 astrocyte markers via qPCR. RESULTS We report structural remodeling of central amygdala microglia and astrocytes during heart failure that affected cell volume, surface area, filament length, and glial branches, resulting overall in somatic swelling and deramification, indicative of a change in glial state. These changes occurred in a time-dependent manner, correlated with the severity of heart failure, and were delayed compared to changes in the hypothalamic paraventricular nucleus. Morphometric changes correlated with elevated mRNA levels of pro-inflammatory cytokines and markers of reactive A1-type astrocytes in the paraventricular nucleus and central amygdala during heart failure. CONCLUSION We provide evidence that in addition to the previously described hypothalamic neuroinflammation implicated in sympathohumoral activation during heart failure, microglia, and astrocytes within the central amygdala also undergo structural remodeling indicative of glial shifts towards pro-inflammatory phenotypes. Thus, our studies suggest that neuroinflammation in the amygdala stands as a novel pathophysiological mechanism and potential therapeutic target that could be associated with emotional and cognitive deficits commonly observed at later stages during the course of heart failure.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA
| | | | | | - Ranjan K Roy
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA
| | - Atit A Patel
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Anne Murphy
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Daniel N Cox
- Neuroscience Institute, Georgia State University, Atlanta, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, USA.
| |
Collapse
|
220
|
Schwartz M, Peralta Ramos JM, Ben-Yehuda H. A 20-Year Journey from Axonal Injury to Neurodegenerative Diseases and the Prospect of Immunotherapy for Combating Alzheimer's Disease. THE JOURNAL OF IMMUNOLOGY 2020; 204:243-250. [PMID: 31907265 DOI: 10.4049/jimmunol.1900844] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
The understanding of the dialogue between the brain and the immune system has undergone dramatic changes over the last two decades, with immense impact on the perception of neurodegenerative diseases, mental dysfunction, and many other brain pathologic conditions. Accumulated results have suggested that optimal function of the brain is dependent on support from the immune system, provided that this immune response is tightly controlled. Moreover, in contrast to the previous prevailing dogma, it is now widely accepted that circulating immune cells are needed for coping with brain pathologies and that their optimal effect is dependent on their type, location, and activity. In this perspective, we describe our own scientific journey, reviewing the milestones in attaining this understanding of the brain-immune axis integrated with numerous related studies by others. We then explain their significance in demonstrating the possibility of harnessing the immune system in a well-controlled manner for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Michal Schwartz
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142; and .,Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | | | - Hila Ben-Yehuda
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
221
|
Ashford BA, Boche D, Cooper-Knock J, Heath PR, Simpson JE, Highley JR. Review: Microglia in motor neuron disease. Neuropathol Appl Neurobiol 2020; 47:179-197. [PMID: 32594542 DOI: 10.1111/nan.12640] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023]
Abstract
Motor Neuron Disease (MND) is a fatal neurodegenerative condition, which is characterized by the selective loss of the upper and lower motor neurons. At the sites of motor neuron injury, accumulation of activated microglia, the primary immune cells of the central nervous system, is commonly observed in both human post mortem studies and animal models of MND. Microglial activation has been found to correlate with many clinical features and importantly, the speed of disease progression in humans. Both anti-inflammatory and pro-inflammatory microglial responses have been shown to influence disease progression in humans and models of MND. As such, microglia could both contribute to and protect against inflammatory mechanisms of pathogenesis in MND. While murine models have characterized the microglial response to MND, these studies have painted a complex and often contradictory picture, indicating a need for further characterization in humans. This review examines the potential role microglia play in MND in human and animal studies. Both the pro-inflammatory and anti-inflammatory responses will be addressed, throughout the course of disease, followed by the potential of microglia as a target in the development of disease-modifying treatments for MND.
Collapse
Affiliation(s)
| | - D Boche
- University of Southampton, Southampton, UK
| | | | - P R Heath
- University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
222
|
Abstract
Microglia are the major immune cells in the central nervous system and play a key role in the normal function of the brain. Microglia exhibit functional diversity, and they control the inflammation in central nervous system through releasing inflammatory cytokine, clearing apoptotic cells via phagocytosis, regulating synaptic plasticity and the formation of neural network by synapse pruning. Recent studies have strongly indicated that the microglial dysfunction is associated with a variety of neuropsychiatric diseases such as depression, which have been termed as "microgliopathy". The emergency of advanced technologies and tools has enabled us to comprehensively understand the role of microglia in physiology and pathology, and growing studies have targetted microglia to explore the treatment of neuropsychiatric diseases. Here, we describe the key progress of microglia research, and review the recent developments in the understanding of the role of microglia in physiology and etiology of depression.
Collapse
|
223
|
Ascenzi P, Bettinelli M, Boffi A, Botta M, De Simone G, Luchinat C, Marengo E, Mei H, Aime S. Rare earth elements (REE) in biology and medicine. RENDICONTI LINCEI-SCIENZE FISICHE E NATURALI 2020. [DOI: 10.1007/s12210-020-00930-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractThis survey reports on topics that were presented at the workshop on “Challenges with Rare Earth Elements. The Periodic Table at work for new Science & Technology” hold at the Academia dei Lincei in November 2019. The herein reported materials refer to presentations dealing with studies and applications of rare earth elements (REE) in several areas of Biology and Medicine. All together they show the tremendous impact REE have in relevant fields of living systems and highlight, on one hand, the still existing knowledge gap for an in-depth understanding of their function in natural systems as well as the very important role they already have in providing innovative scientific and technological solutions in a number of bio-medical areas and in fields related to the assessment of the origin of food and on their manufacturing processes. On the basis of the to-date achievements one expects that new initiatives will bring, in a not too far future, to a dramatic increase of our understanding of the REE involvement in living organisms as well as a ramp-up in the exploitation of the peculiar properties of REE for the design of novel applications in diagnostic procedures and in the set-up of powerful medical devices. This scenario calls the governmental authorities for new responsibilities to guarantee a continuous availability of REE to industry and research labs together with providing support to activities devoted to their recovery/recycling.
Collapse
|
224
|
Maternal Immune Activation Sensitizes Male Offspring Rats to Lipopolysaccharide-Induced Microglial Deficits Involving the Dysfunction of CD200-CD200R and CX3CL1-CX3CR1 Systems. Cells 2020; 9:cells9071676. [PMID: 32664639 PMCID: PMC7407118 DOI: 10.3390/cells9071676] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023] Open
Abstract
Early life challenges resulting from maternal immune activation (MIA) may exert persistent effects on the offspring, including the development of psychiatric disorders, such as schizophrenia. Recent evidence has suggested that the adverse effects of MIA may be mediated by neuron-microglia crosstalk, particularly CX3CL1-CX3CR1 and CD200-CD200R dyads. Therefore, the present study assessed the behavioural parameters resembling schizophrenia-like symptoms in the adult male offspring of Sprague-Dawley rats that were exposed to MIA and to an additional acute lipopolysaccharide (LPS) challenge in adulthood, according to the "two-hit" hypothesis of schizophrenia. Simultaneously, we aimed to clarify the role of the CX3CL1-CX3CR1 and CD200-CD200R axes and microglial reactivity in the brains of adult offspring subjected to MIA and the "second hit" wit LPS. In the present study, MIA generated a range of behavioural changes in the adult male offspring, including increased exploratory activity and anxiety-like behaviours. The most intriguing finding was observed in the prepulse inhibition (PPI) test, where the deficit in the sensorimotor gating was age-dependent and present only in part of the rats. We were able to distinguish the occurrence of two groups: responsive and non-responsive (without the deficit). Concurrently, based on the results of the biochemical studies, MIA disrupted mainly the CD200-CD200R system, while the changes of the CX3CL1-CX3CR1 axis were less evident in the frontal cortex of adult non-responsive offspring. MIA markedly affected the immune regulators of the CD200-CD200R pathway as we observed an increase in cortical IL-6 release in the responsive group and IL-4 in the non-responsive offspring. Importantly, the "second hit" generated disturbances at the behavioural and biochemical levels mostly in the non-responsive adult animals. Those offspring were characterized both by disturbed PPI and "priming" microglia. Altogether, the exposure to MIA altered the immunomodulatory mechanisms, including the CD200-CD200R axis, in the brain and sensitized animals to subsequent immunological challenges, leading to the manifestation of schizophrenia-like alterations.
Collapse
|
225
|
Masuda T, Amann L, Sankowski R, Staszewski O, Lenz M, D Errico P, Snaidero N, Costa Jordão MJ, Böttcher C, Kierdorf K, Jung S, Priller J, Misgeld T, Vlachos A, Meyer-Luehmann M, Knobeloch KP, Prinz M. Novel Hexb-based tools for studying microglia in the CNS. Nat Immunol 2020; 21:802-815. [PMID: 32541832 DOI: 10.1038/s41590-020-0707-4] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 05/07/2020] [Indexed: 12/26/2022]
Abstract
Microglia and central nervous system (CNS)-associated macrophages (CAMs), such as perivascular and meningeal macrophages, are implicated in virtually all diseases of the CNS. However, little is known about their cell-type-specific roles in the absence of suitable tools that would allow for functional discrimination between the ontogenetically closely related microglia and CAMs. To develop a new microglia gene targeting model, we first applied massively parallel single-cell analyses to compare microglia and CAM signatures during homeostasis and disease and identified hexosaminidase subunit beta (Hexb) as a stably expressed microglia core gene, whereas other microglia core genes were substantially downregulated during pathologies. Next, we generated HexbtdTomato mice to stably monitor microglia behavior in vivo. Finally, the Hexb locus was employed for tamoxifen-inducible Cre-mediated gene manipulation in microglia and for fate mapping of microglia but not CAMs. In sum, we provide valuable new genetic tools to specifically study microglia functions in the CNS.
Collapse
Affiliation(s)
- Takahiro Masuda
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| | - Lukas Amann
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ori Staszewski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Paolo D Errico
- Department of Neurology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nicolas Snaidero
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | | | - Chotima Böttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZNE and BIH, Berlin, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center-University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus-Peter Knobeloch
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
226
|
Sebastian Monasor L, Müller SA, Colombo AV, Tanrioever G, König J, Roth S, Liesz A, Berghofer A, Piechotta A, Prestel M, Saito T, Saido TC, Herms J, Willem M, Haass C, Lichtenthaler SF, Tahirovic S. Fibrillar Aβ triggers microglial proteome alterations and dysfunction in Alzheimer mouse models. eLife 2020; 9:54083. [PMID: 32510331 PMCID: PMC7279888 DOI: 10.7554/elife.54083] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 05/02/2020] [Indexed: 12/20/2022] Open
Abstract
Microglial dysfunction is a key pathological feature of Alzheimer's disease (AD), but little is known about proteome-wide changes in microglia during the course of AD and their functional consequences. Here, we performed an in-depth and time-resolved proteomic characterization of microglia in two mouse models of amyloid β (Aβ) pathology, the overexpression APPPS1 and the knock-in APP-NL-G-F (APP-KI) model. We identified a large panel of Microglial Aβ Response Proteins (MARPs) that reflect heterogeneity of microglial alterations during early, middle and advanced stages of Aβ deposition and occur earlier in the APPPS1 mice. Strikingly, the kinetic differences in proteomic profiles correlated with the presence of fibrillar Aβ, rather than dystrophic neurites, suggesting that fibrillar Aβ may trigger the AD-associated microglial phenotype and the observed functional decline. The identified microglial proteomic fingerprints of AD provide a valuable resource for functional studies of novel molecular targets and potential biomarkers for monitoring AD progression or therapeutic efficacy. Alzheimer’s disease is a progressive, irreversible brain disorder. Patients with Alzheimer’s have problems with memory and other mental skills, which lead to more severe cognitive decline and, eventually, premature death. This is due to increasing numbers of nerve cells in the brain dying over time. A distinctive feature of Alzheimer’s is the abnormally high accumulation of a protein called amyloid-β, which forms distinctive clumps in the brain termed ‘plaques’. The brain has a type of cells called the microglia that identify infections, toxic material and damaged cells, and prevent these from building up by clearing them away. In Alzheimer’s disease, however, the microglia do not work properly, which is thought to contribute to the accumulation of amyloid-β plaques. This means that people with mutations in the genes important for the microglia activity are also at higher risk of developing the disease. Although problems with the microglia play an important role in Alzheimer’s, researchers still do not fully understand why microglia stop working in the first place. It is also not known exactly when and how the microglia change as Alzheimer’s disease progresses. To unravel this mystery, Sebastian Monasor, Müller et al. carried out a detailed study of the molecular ‘fingerprints’ of microglia at each key stage of Alzheimer’s disease. The experiments used microglia cells from two different strains of genetically altered mice, both of which develop the hallmarks of Alzheimer’s disease, including amyloid-β plaques, at similar rates. Analysis of the proteins in microglia cells from both strains revealed distinctive, large-scale changes corresponding to successive stages of the disease – reflecting the gradual accumulation of plaques. Obvious defects in microglia function also appeared soon after plaques started to build up. Microscopy imaging of the brain tissue showed that although amyloid-β plaques appeared at the same time, they looked different in each mouse strain. In one, plaques were more compact, while in the other, plaques appeared ‘fluffier’, like cotton wool. In mice with more compacted plaques, microglia recognized the plaques earlier and stopped working sooner, suggesting that plaque structure and microglia defects could be linked. These results shed new light on the role of microglia and their changing protein ‘signals’ during the different stages of Alzheimer’s disease. In the future, this information could help identify people at risk for the disease, so that they can be treated as soon as possible, and to design new therapies to make microglia work again.
Collapse
Affiliation(s)
- Laura Sebastian Monasor
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Graduate School of Systemic Neuroscience, Ludwig-Maximilians-University, Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | | - Gaye Tanrioever
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.,Department of Cellular Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Jasmin König
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Faculty of Chemistry, Technical University of Munich, Garching, Germany
| | - Stefan Roth
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU, Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Anna Berghofer
- Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University, Munich, Germany
| | - Anke Piechotta
- Department of Molecular Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | - Matthias Prestel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU, Munich, Germany
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science Institute, Wako, Japan.,Department of Neurocognitive Science, Nagoya City University Graduate School of Medical Science, Nagoya, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science Institute, Wako, Japan
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Willem
- Biomedical Center (BMC), Ludwig-Maximilians Universität München, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Biomedical Center (BMC), Ludwig-Maximilians Universität München, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University, Munich, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
227
|
Machhi J, Kevadiya BD, Muhammad IK, Herskovitz J, Olson KE, Mosley RL, Gendelman HE. Harnessing regulatory T cell neuroprotective activities for treatment of neurodegenerative disorders. Mol Neurodegener 2020; 15:32. [PMID: 32503641 PMCID: PMC7275301 DOI: 10.1186/s13024-020-00375-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence demonstrates that adaptive immunity influences the pathobiology of neurodegenerative disorders. Misfolded aggregated self-proteins can break immune tolerance leading to the induction of autoreactive effector T cells (Teffs) with associated decreases in anti-inflammatory neuroprotective regulatory T cells (Tregs). An imbalance between Teffs and Tregs leads to microglial activation, inflammation and neuronal injury. The cascade of such a disordered immunity includes the drainage of the aggregated protein antigens into cervical lymph nodes serving to amplify effector immune responses. Both preclinical and clinical studies demonstrate transformation of this altered immunity for therapeutic gain. We posit that the signs and symptoms of common neurodegenerative disorders such as Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, and stroke can be attenuated by boosting Treg activities.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Bhavesh D. Kevadiya
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Radiology, School of Medicine, Stanford University, Palo Alto, 94304 USA
| | - Ijaz Khan Muhammad
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pharmacy, University of Swabi, Anbar Swabi, 23561 Pakistan
| | - Jonathan Herskovitz
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| |
Collapse
|
228
|
Sciara AN, Beasley B, Crawford JD, Anderson EP, Carrasco T, Zheng S, Ordway GA, Chandley MJ. Neuroinflammatory Gene Expression Alterations in Anterior Cingulate Cortical White and Gray Matter of Males With Autism Spectrum Disorder. Autism Res 2020; 13:870-884. [PMID: 32129578 PMCID: PMC7540672 DOI: 10.1002/aur.2284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 01/26/2020] [Accepted: 02/17/2020] [Indexed: 02/06/2023]
Abstract
Evidence for putative pathophysiological mechanisms of autism spectrum disorder (ASD), including peripheral inflammation, blood-brain barrier disruption, white matter alterations, and abnormal synaptic overgrowth, indicate a possible involvement of neuroinflammation in the disorder. Neuroinflammation plays a role in the development and maintenance of the dendritic spines involved in glutamatergic and GABAergic neurotransmission, and also influences blood-brain permeability. Cytokines released from microglia can impact the length, location or organization of dendritic spines on excitatory and inhibitory cells as well as recruit and impact glial cell function around the neurons. In this study, gene expression levels of anti- and pro-inflammatory signaling molecules, as well as oligodendrocyte and astrocyte marker proteins, were measured in both gray and white matter tissue in the anterior cingulate cortex from ASD and age-matched typically developing (TD) control brain donors, ranging from ages 4 to 37 years. Expression levels of the pro-inflammatory gene, HLA-DR, were significantly reduced in gray matter and expression levels of the anti-inflammatory gene MRC1 were significantly elevated in white matter from ASD donors as compared to TD donors, but neither retained statistical significance after correction for multiple comparisons. Modest trends toward differences in expression levels were also observed for the pro-inflammatory (CD68, IL1β) and anti-inflammatory genes (IGF1, IGF1R) comparing ASD donors to TD donors. The direction of gene expression changes comparing ASD to TD donors did not reveal consistent findings implicating an elevated pro- or anti-inflammatory state in ASD. However, altered expression of pro- and anti-inflammatory gene expression indicates some involvement of neuroinflammation in ASD. Autism Res 2020, 13: 870-884. © 2020 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: The anterior cingulate cortex is an integral brain region in modulating social behaviors including nonverbal communication. The study found that inflammatory gene expression levels were altered in this brain region. We hypothesize that the inflammatory changes in this area could impact neuronal function. The finding has future implications in using these molecular markers to identify potential environmental exposures and distinct cell differences in autism.
Collapse
Affiliation(s)
- Aubrey N. Sciara
- Department of Biological SciencesEast Tennessee State UniversityJohnson CityTennessee
| | - Brooke Beasley
- Department of Health SciencesEast Tennessee State UniversityJohnson CityTN
| | - Jessica D. Crawford
- Department of Biomedical SciencesEast Tennessee State UniversityJohnson CityTN
| | - Emma P. Anderson
- Department of Health SciencesEast Tennessee State UniversityJohnson CityTN
| | - Tiffani Carrasco
- Department of Health SciencesEast Tennessee State UniversityJohnson CityTN
| | - Shimin Zheng
- Department of Biostatistics and EpidemiologyEast Tennessee State UniversityJohnson CityTN
| | - Gregory A. Ordway
- Department of Biomedical SciencesEast Tennessee State UniversityJohnson CityTN
- Department of Psychiatry and Behavioral SciencesEast Tennessee State University, Johnson CityJohnson CityTN
| | | |
Collapse
|
229
|
Prinz M, Jung S, Priller J. Microglia Biology: One Century of Evolving Concepts. Cell 2020; 179:292-311. [PMID: 31585077 DOI: 10.1016/j.cell.2019.08.053] [Citation(s) in RCA: 847] [Impact Index Per Article: 169.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/08/2019] [Accepted: 08/27/2019] [Indexed: 01/05/2023]
Abstract
Microglia were first recognized as a distinct cell population in the CNS one century ago. For a long time, they were primarily considered to be phagocytes responsible for removing debris during CNS development and disease. More recently, advances in imaging and genetics and the advent of single-cell technologies provided new insights into the much more complex and fascinating biology of microglia. The ontogeny of microglia was identified, and their functions in health and disease were better defined. Although many questions about microglia and their roles in human diseases remain unanswered, the prospect of targeting microglia for the treatment of neurological and psychiatric disorders is tantalizing.
Collapse
Affiliation(s)
- Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany; DZNE and BIH, Berlin, Germany; University of Edinburgh and UK DRI, Edinburgh, UK.
| |
Collapse
|
230
|
Yang Y, Zhang Z. Microglia and Wnt Pathways: Prospects for Inflammation in Alzheimer's Disease. Front Aging Neurosci 2020; 12:110. [PMID: 32477095 PMCID: PMC7241259 DOI: 10.3389/fnagi.2020.00110] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/30/2020] [Indexed: 01/27/2023] Open
Abstract
Alzheimer’s disease (AD) has been a major health issue for more than one century since it was first reported in 1906. As one of the most common neurodegenerative diseases, AD is characterized by the presence of senile plaques and neurofibrillary tangles (NFTs) in the affected brain area. Microglia are the major regulators of neuroinflammation in the brain, and neuroinflammation has become recognized as the core pathophysiological process of various neurodegenerative diseases. In the central nervous system (CNS), microglia play a dual role in AD development. For one thing, they degrade amyloid β (Aβ) to resist its deposition; for another, microglia release pro-inflammatory and inflammatory factors, contributing to neuroinflammation as well as the spreading of Aβ and tau pathology. Wnt pathways are important regulators of cell fate and cell activities. The dysregulation of Wnt pathways is responsible for both abnormal tau phosphorylation and synaptic loss in AD. Recent studies have also confirmed the regulatory effect of Wnt signaling on microglial inflammation. Thus, the study of microglia, Wnt pathways, and their possible interactions may open up a new direction for understanding the mechanisms of neuroinflammation in AD. In this review, we summarize the functions of microglia and Wnt pathways and their roles in AD in order to provide new ideas for understanding the pathogenesis of AD.
Collapse
Affiliation(s)
- Yunying Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
231
|
Rayaprolu S, Gao T, Xiao H, Ramesha S, Weinstock LD, Shah J, Duong DM, Dammer EB, Webster JA, Lah JJ, Wood LB, Betarbet R, Levey AI, Seyfried NT, Rangaraju S. Flow-cytometric microglial sorting coupled with quantitative proteomics identifies moesin as a highly-abundant microglial protein with relevance to Alzheimer's disease. Mol Neurodegener 2020; 15:28. [PMID: 32381088 PMCID: PMC7206797 DOI: 10.1186/s13024-020-00377-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/24/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Proteomic characterization of microglia provides the most proximate assessment of functionally relevant molecular mechanisms of neuroinflammation. However, microglial proteomics studies have been limited by low cellular yield and contamination by non-microglial proteins using existing enrichment strategies. METHODS We coupled magnetic-activated cell sorting (MACS) and fluorescence activated cell sorting (FACS) of microglia with tandem mass tag-mass spectrometry (TMT-MS) to obtain a highly-pure microglial proteome and identified a core set of highly-abundant microglial proteins in adult mouse brain. We interrogated existing human proteomic data for Alzheimer's disease (AD) relevance of highly-abundant microglial proteins and performed immuno-histochemical and in-vitro validation studies. RESULTS Quantitative multiplexed proteomics by TMT-MS of CD11b + MACS-enriched (N = 5 mice) and FACS-isolated (N = 5 mice), from adult wild-type mice, identified 1791 proteins. A total of 203 proteins were highly abundant in both datasets, representing a core-set of highly abundant microglial proteins. In addition, we found 953 differentially enriched proteins comparing MACS and FACS-based approaches, indicating significant differences between both strategies. The FACS-isolated microglia proteome was enriched with cytosolic, endoplasmic reticulum, and ribosomal proteins involved in protein metabolism and immune system functions, as well as an abundance of canonical microglial proteins. Conversely, the MACS-enriched microglia proteome was enriched with mitochondrial and synaptic proteins and higher abundance of neuronal, oligodendrocytic and astrocytic proteins. From the 203 consensus microglial proteins with high abundance in both datasets, we confirmed microglial expression of moesin (Msn) in wild-type and 5xFAD mouse brains as well as in human AD brains. Msn expression is nearly exclusively found in microglia that surround Aβ plaques in 5xFAD brains. In in-vitro primary microglial studies, Msn silencing by siRNA decreased Aβ phagocytosis and increased lipopolysaccharide-induced production of the pro-inflammatory cytokine, tumor necrosis factor (TNF). In network analysis of human brain proteomic data, Msn was a hub protein of an inflammatory co-expression module positively associated with AD neuropathological features and cognitive dysfunction. CONCLUSIONS Using FACS coupled with TMT-MS as the method of choice for microglial proteomics, we define a core set of highly-abundant adult microglial proteins. Among these, we validate Msn as highly-abundant in plaque-associated microglia with relevance to human AD.
Collapse
Affiliation(s)
- Sruti Rayaprolu
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
| | - Tianwen Gao
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
- Xiangya Hospital, Central South University, Changsha, 410008 Hunan China
| | - Hailian Xiao
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
| | - Supriya Ramesha
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
| | - Laura D. Weinstock
- Parker H. Petit Institute for Bioengineering and Bioscience, Wallace H. Coulter Department of Biomedical Engineering, and Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Jheel Shah
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
| | - Duc M. Duong
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
- Department of Biochemistry, Emory University, Atlanta, GA 30322 USA
| | - Eric B. Dammer
- School of Medicine, Emory University, Atlanta, GA 30322 USA
| | - James A. Webster
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
| | - James J. Lah
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
| | - Levi B. Wood
- Parker H. Petit Institute for Bioengineering and Bioscience, Wallace H. Coulter Department of Biomedical Engineering, and Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Ranjita Betarbet
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
| | - Allan I. Levey
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
| | - Nicholas T. Seyfried
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
- Department of Biochemistry, Emory University, Atlanta, GA 30322 USA
| | - Srikant Rangaraju
- Department of Neurology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322 USA
| |
Collapse
|
232
|
Hatcher A, Yu K, Meyer J, Aiba I, Deneen B, Noebels JL. Pathogenesis of peritumoral hyperexcitability in an immunocompetent CRISPR-based glioblastoma model. J Clin Invest 2020; 130:2286-2300. [PMID: 32250339 PMCID: PMC7190940 DOI: 10.1172/jci133316] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/17/2020] [Indexed: 12/19/2022] Open
Abstract
Seizures often herald the clinical appearance of gliomas or appear at later stages. Dissecting their precise evolution and cellular pathogenesis in brain malignancies could inform the development of staged therapies for these highly pharmaco-resistant epilepsies. Studies in immunodeficient xenograft models have identified local interneuron loss and excess glial glutamate release as chief contributors to network disinhibition, but how hyperexcitability in the peritumoral microenvironment evolves in an immunocompetent brain is unclear. We generated gliomas in WT mice via in utero deletion of key tumor suppressor genes and serially monitored cortical epileptogenesis during tumor infiltration with in vivo electrophysiology and GCAMP7 calcium imaging, revealing a reproducible progression from hyperexcitability to convulsive seizures. Long before seizures, coincident with loss of inhibitory cells and their protective scaffolding, gain of glial glutamate antiporter xCT expression, and reactive astrocytosis, we detected local Iba1+ microglial inflammation that intensified and later extended far beyond tumor boundaries. Hitherto unrecognized episodes of cortical spreading depolarization that arose frequently from the peritumoral region may provide a mechanism for transient neurological deficits. Early blockade of glial xCT activity inhibited later seizures, and genomic reduction of host brain excitability by deleting MapT suppressed molecular markers of epileptogenesis and seizures. Our studies confirmed xenograft tumor-driven pathobiology and revealed early and late components of tumor-related epileptogenesis in a genetically tractable, immunocompetent mouse model of glioma, allowing the complex dissection of tumor versus host pathogenic seizure mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | - Jeffrey L. Noebels
- Department of Neuroscience
- Department of Neurology, and
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
233
|
Plastira I, Bernhart E, Joshi L, Koyani CN, Strohmaier H, Reicher H, Malle E, Sattler W. MAPK signaling determines lysophosphatidic acid (LPA)-induced inflammation in microglia. J Neuroinflammation 2020; 17:127. [PMID: 32326963 PMCID: PMC7178949 DOI: 10.1186/s12974-020-01809-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
Background In the extracellular environment, lysophosphatidic acid (LPA) species are generated via autotaxin (ATX)-mediated hydrolysis of lysophospholipid precursors. Members of the LPA family are potent lipid mediators transmitting signals via six different G protein-coupled LPA receptors (LPAR1-6). The LPA signaling axis is indispensable for brain development and function of the nervous system; however, during damage of the central nervous system, LPA levels can increase and aberrant signaling events counteract brain function. Here, we investigated regulation of the ATX/LPA/LPAR axis in response to lipopolysaccharide-induced systemic inflammation in mice and potential neurotoxic polarization programs in LPA-activated primary murine microglia. Methods In vivo, LPAR1-6 expression was established by qPCR in whole murine brain homogenates and in FACS-sorted microglia. ELISAs were used to quantitate LPA concentrations in the brain and cyto-/chemokine secretion from primary microglia in vitro. Transcription factor phosphorylation was analyzed by immunoblotting, and plasma membrane markers were analyzed by flow cytometry. We used MAPK inhibitors to study signal integration by the JNK, p38, and ERK1/2 branches in response to LPA-mediated activation of primary microglia. Results Under acute and chronic inflammatory conditions, we observed a significant increase in LPA concentrations and differential regulation of LPAR, ATX (encoded by ENPP2), and cytosolic phospholipase A2 (encoded by PLA2G4A) gene expression in the brain and FACS-sorted microglia. During pathway analyses in vitro, the use of specific MAPK antagonists (SP600125, SB203580, and PD98059) revealed that JNK and p38 inhibition most efficiently attenuated LPA-induced phosphorylation of proinflammatory transcription factors (STAT1 and -3, p65, and c-Jun) and secretion of IL-6 and TNFα. All three inhibitors decreased LPA-mediated secretion of IL-1β, CXCL10, CXCL2, and CCL5. The plasma membrane marker CD40 was solely inhibited by SP600125 while all three inhibitors affected expression of CD86 and CD206. All MAPK antagonists reduced intracellular COX-2 and Arg1 as well as ROS and NO formation, and neurotoxicity of microglia-conditioned media. Conclusion In the present study, we show that systemic inflammation induces aberrant ATX/LPA/LPAR homeostasis in the murine brain. LPA-mediated polarization of primary microglia via MAPK-dependent pathways induces features reminiscent of a neurotoxic phenotype.
Collapse
Affiliation(s)
- Ioanna Plastira
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Eva Bernhart
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Lisha Joshi
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Chintan N Koyani
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria.,Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Heimo Strohmaier
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Helga Reicher
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria
| | - Wolfgang Sattler
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010, Graz, Austria. .,Center for Explorative Lipidomics, BioTechMed, Graz, Austria.
| |
Collapse
|
234
|
Burns M, Schulz AR, Kunkel D, Hönig M, Warth S, Bengsch B, Burns T, Reinhardt J, Grützkau A, Yaspo ML, Sodenkamp J, Hoffmann U, Mei HE. Mass Cytometry-A Tool for the Curious: Networking in Berlin. Cytometry A 2020; 97:764-767. [PMID: 32298052 DOI: 10.1002/cyto.a.24015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 11/09/2022]
Affiliation(s)
| | | | - Désirée Kunkel
- Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Manfred Hönig
- Universität Ulm, Medizinische Fakultät, Ulm, Germany
| | - Sarah Warth
- Universität Ulm, Medizinische Fakultät, Ulm, Germany
| | - Bertram Bengsch
- Department of Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Tyler Burns
- DRFZ Berlin, a Leibniz Institute, Berlin, Germany
| | - Julia Reinhardt
- Technische Universität Dresden, Center for Regenerative Therapies Dresden, Dresden, Germany
| | | | | | - Jan Sodenkamp
- TranslaTUM, Technische Universität München, Munich, Germany
| | - Ute Hoffmann
- DRFZ Berlin, a Leibniz Institute, Berlin, Germany
| | - Henrik E Mei
- DRFZ Berlin, a Leibniz Institute, Berlin, Germany
| |
Collapse
|
235
|
Norton SE, Leman JKH, Khong T, Spencer A, Fazekas de St Groth B, McGuire HM, Kemp RA. Brick plots: an intuitive platform for visualizing multiparametric immunophenotyped cell clusters. BMC Bioinformatics 2020; 21:145. [PMID: 32293253 PMCID: PMC7158154 DOI: 10.1186/s12859-020-3469-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/24/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The advent of mass cytometry has dramatically increased the parameter limit for immunological analysis. New approaches to analysing high parameter cytometry data have been developed to ease analysis of these complex datasets. Many of these methods assign cells into population clusters based on protein expression similarity. RESULTS Here we introduce an additional method, termed Brick plots, to visualize these cluster phenotypes in a simplified and intuitive manner. The Brick plot method generates a two-dimensional barcode that displays the phenotype of each cluster in relation to the entire dataset. We show that Brick plots can be used to visualize complex mass cytometry data, both from fundamental research and clinical trials, as well as flow cytometry data. CONCLUSION Brick plots represent a new approach to visualize complex immunological data in an intuitive manner.
Collapse
Affiliation(s)
- Samuel E Norton
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Julia K H Leman
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Tiffany Khong
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital-Monash University, Melbourne, VIC, Australia
- Malignant Hematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital-Monash University, Melbourne, VIC, Australia
- Malignant Hematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Barbara Fazekas de St Groth
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, Australia
- Discipline of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia; Charles Perkins Centre, University of Sydney, Sydney, Australia
| | - Helen M McGuire
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, Australia.
- Discipline of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Australia; Charles Perkins Centre, University of Sydney, Sydney, Australia.
| | - Roslyn A Kemp
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
236
|
Benmamar-Badel A, Owens T, Wlodarczyk A. Protective Microglial Subset in Development, Aging, and Disease: Lessons From Transcriptomic Studies. Front Immunol 2020; 11:430. [PMID: 32318054 PMCID: PMC7147523 DOI: 10.3389/fimmu.2020.00430] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/25/2020] [Indexed: 12/25/2022] Open
Abstract
Microglial heterogeneity has been the topic of much discussion in the scientific community. Elucidation of their plasticity and adaptability to disease states triggered early efforts to characterize microglial subsets. Over time, their phenotypes, and later on their homeostatic signature, were revealed, through the use of increasingly advanced transcriptomic techniques. Recently, an increasing number of these "microglial signatures" have been reported in various homeostatic and disease contexts. Remarkably, many of these states show similar overlapping microglial gene expression patterns, both in homeostasis and in disease or injury. In this review, we integrate information from these studies, and we propose a unique subset, for which we introduce a core signature, based on our own research and reports from the literature. We describe that this subset is found in development and in normal aging as well as in diverse diseases. We discuss the functions of this subset as well as how it is induced.
Collapse
Affiliation(s)
- Anouk Benmamar-Badel
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
- Department of Neurology, Slagelse Hospital, Institute of Regional Health Research, Slagelse, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| |
Collapse
|
237
|
von Ehr A, Attaai A, Neidert N, Potru PS, Ruß T, Zöller T, Spittau B. Inhibition of Microglial TGFβ Signaling Increases Expression of Mrc1. Front Cell Neurosci 2020; 14:66. [PMID: 32296307 PMCID: PMC7137652 DOI: 10.3389/fncel.2020.00066] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/04/2020] [Indexed: 01/05/2023] Open
Abstract
Microglia are constantly surveying their microenvironment and rapidly react to impairments by changing their morphology, migrating toward stimuli and adopting gene expression profiles characterizing their activated state. The increased expression of the M2-like marker Mannose receptor 1 (Mrc1), which is also referred to as CD206, in microglia has been reported after M2-like activation in vitro and in vivo. Mrc1 is a 175-kDa transmembrane pattern recognition receptor which binds a variety of carbohydrates and is involved in the pinocytosis and the phagocytosis of immune cells, including microglia, and thought to contribute to a neuroprotective microglial phenotype. Here we analyzed the effects of TGFβ signaling on Mrc1 expression in microglia in vivo and in vitro. Using C57BL/6 wild type and Cx3cr1CreERT2:R26-YFP:Tgfbr2fl/fl mice-derived microglia, we show that the silencing of TGFβ signaling results in the upregulation of Mrc1, whereas recombinant TGFβ1 induced the delayed downregulation of Mrc1. Furthermore, chromatin immunoprecipitation experiments provided evidence that Mrc1 is not a direct Smad2/Smad4 target gene in microglia. Altogether our data indicate that the changes in Mrc1 expression after the activation or the silencing of microglial TGFβ signaling are likely to be mediated by modifications of the secondary intracellular signaling events influenced by TGFβ signaling.
Collapse
Affiliation(s)
- Alexander von Ehr
- Department of Molecular Embryology, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Abdelraheim Attaai
- Department of Molecular Embryology, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany.,Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Nicolas Neidert
- Department of Molecular Embryology, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | | | - Tamara Ruß
- Institute of Anatomy, University of Rostock, Rostock, Germany
| | - Tanja Zöller
- Institute of Anatomy, University of Rostock, Rostock, Germany
| | - Björn Spittau
- Institute of Anatomy, University of Rostock, Rostock, Germany.,Centre for Translational Neurosciences Rostock, Rostock, Germany
| |
Collapse
|
238
|
Gao J, Littman R, Diamante G, Xiao X, Ahn IS, Yang X, Cole TA, Tontonoz P. Therapeutic IDOL Reduction Ameliorates Amyloidosis and Improves Cognitive Function in APP/PS1 Mice. Mol Cell Biol 2020; 40:e00518-19. [PMID: 31964754 PMCID: PMC7108818 DOI: 10.1128/mcb.00518-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/09/2019] [Accepted: 01/11/2020] [Indexed: 01/10/2023] Open
Abstract
Brain lipoprotein receptors have been shown to regulate the metabolism of ApoE and β-amyloid (Aβ) and are potential therapeutic targets for Alzheimer's disease (AD). Previously, we identified E3 ubiquitin ligase IDOL as a negative regulator of brain lipoprotein receptors. Genetic ablation of Idol increases low-density lipoprotein receptor protein levels, which facilitates Aβ uptake and clearance by microglia. In this study, we utilized an antisense oligonucleotide (ASO) to reduce IDOL expression therapeutically in the brains of APP/PS1 male mice. ASO treatment led to decreased Aβ pathology and improved spatial learning and memory. Single-cell transcriptomic analysis of hippocampus revealed that IDOL inhibition upregulated lysosomal/phagocytic genes in microglia. Furthermore, clustering of microglia revealed that IDOL-ASO treatment shifted the composition of the microglia population by increasing the prevalence of disease-associated microglia. Our results suggest that reducing IDOL expression in the adult brain promotes the phagocytic clearance of Aβ and ameliorates Aβ-dependent pathology. Pharmacological inhibition of IDOL activity in the brain may represent a therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Jie Gao
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Russell Littman
- Department of Integrative Biology and Physiology, University of California-Los Angeles, Los Angeles, California, USA
- Bioinformatics Interdepartmental Program, University of California-Los Angeles, Los Angeles, California, USA
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California-Los Angeles, Los Angeles, California, USA
| | - Xu Xiao
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - In Sook Ahn
- Department of Integrative Biology and Physiology, University of California-Los Angeles, Los Angeles, California, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California-Los Angeles, Los Angeles, California, USA
- Bioinformatics Interdepartmental Program, University of California-Los Angeles, Los Angeles, California, USA
- Institute for Computational and Quantitative Biosciences, University of California-Los Angeles, Los Angeles, California, USA
| | - Tracy A Cole
- Central Nervous System Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| |
Collapse
|
239
|
Affiliation(s)
- Josef Priller
- Department of Neuropsychiatry, Charité and BIH, Berlin, Germany. .,DZNE, Berlin, Germany.,University of Edinburgh and UK DRI, Edinburgh, UK
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany. .,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation, Freiburg, Germany
| |
Collapse
|
240
|
Süß P, Hoffmann A, Rothe T, Ouyang Z, Baum W, Staszewski O, Schett G, Prinz M, Krönke G, Glass CK, Winkler J, Schlachetzki JCM. Chronic Peripheral Inflammation Causes a Region-Specific Myeloid Response in the Central Nervous System. Cell Rep 2020; 30:4082-4095.e6. [PMID: 32209470 DOI: 10.1016/j.celrep.2020.02.109] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 01/15/2020] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
Systemic immune dysregulation contributes to the development of neuropsychiatric and neurodegenerative diseases. The precise effect of chronic peripheral immune stimulation on myeloid cells across anatomical brain regions is unclear. Here, we demonstrate brain-region-specific differences in myeloid responses induced by chronic peripheral inflammation. This shift in the myeloid compartment is associated with the appearance of an inflammatory myeloid subpopulation in the cortex, striatum, and thalamus accompanied by regional transcriptomic fingerprints that include induction of chemokines, complement factors, and endothelial adhesion molecules. In contrast, myeloid immune responses within the hippocampus and cerebellum are subtle or absent. Treatment with the anti-tumor necrosis factor α (anti-TNF-α) antibody infliximab ablates the region-specific inflammatory response. A region-specific myeloid cell response to chronic peripheral inflammation is observed in postmortem brains from individuals with rheumatoid arthritis. Our data suggest that chronic peripheral inflammation has heterogeneous effects on the brain, as evidenced by the spectrum of myeloid cell responses observed across brain regions.
Collapse
Affiliation(s)
- Patrick Süß
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany
| | - Alana Hoffmann
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Tobias Rothe
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie, 91054 Erlangen, Germany
| | - Zhengyu Ouyang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wolfgang Baum
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Ori Staszewski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie, 91054 Erlangen, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany; Signalling Research Centres for BIOSS and CIBSS, University of Freiburg, 79104 Freiburg im Breisgau, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Deutsches Zentrum für Immuntherapie, 91054 Erlangen, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jürgen Winkler
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Johannes C M Schlachetzki
- Department of Molecular Neurology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital Erlangen, 91054 Erlangen, Germany; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
241
|
Erny D, Prinz M. How microbiota shape microglial phenotypes and epigenetics. Glia 2020; 68:1655-1672. [PMID: 32181523 DOI: 10.1002/glia.23822] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/17/2022]
Abstract
Among the myeloid cells in the central nervous system (CNS) microglia are the main representatives of the innate immune system. Microglial fulfil tasks beyond phagocytosing debris and host defense against invading microorganism. During brain development microglia guide for example neurons for proper CNS formation, in adulthood they maintain tissue homeostasis and in aging microglia may become pro-inflammatory and finally exhausted. Recently, several endogenous and exogenous factors were identified that essentially shape the microglial phenotype during both steady-state and pathological conditions. On the one hand, microglia receive inputs from CNS-endogenous sources for example, via crosstalk with other glial cells and neurons but on the other hand microglia are also highly modulated by external signals. Among them, host microbiota-the host's resident bacteria-are vital regulators of the CNS innate immune system. This review summarizes key extrinsic and intrinsic factors, with special focus on the host microbiota, that essentially influence microglia functions and states during development, homeostasis, and disease.
Collapse
Affiliation(s)
- Daniel Erny
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
242
|
Murgoci AN, Duhamel M, Raffo-Romero A, Mallah K, Aboulouard S, Lefebvre C, Kobeissy F, Fournier I, Zilkova M, Maderova D, Cizek M, Cizkova D, Salzet M. Location of neonatal microglia drives small extracellular vesicles content and biological functions in vitro. J Extracell Vesicles 2020; 9:1727637. [PMID: 32158520 PMCID: PMC7049881 DOI: 10.1080/20013078.2020.1727637] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/19/2022] Open
Abstract
Combining proteomics and systems biology approaches, we demonstrate that neonatal microglial cells derived from two different CNS locations, cortex and spinal cord, and cultured in vitro displayed different phenotypes upon different physiological or pathological conditions. These cells also exhibited greater variability in terms of cellular and small extracellular vesicles (sEVs) protein content and levels. Bioinformatic data analysis showed that cortical microglia exerted anti-inflammatory and neurogenesis/tumorigenesis properties, while the spinal cord microglia were more inflammatory. Interestingly, while both sEVs microglia sources enhanced growth of DRGs processes, only the spinal cord-derived sEVs microglia under LPS stimulation significantly attenuated glioma proliferation. These results were confirmed using the neurite outgrowth assay on DRGs cells and glioma proliferation analysis in 3D spheroid cultures. Results from these in vitro assays suggest that the microglia localized at different CNS regions can ensure different biological functions. Together, this study indicates that neonatal microglia locations regulate their physiological and pathological functional fates and could affect the high prevalence of brain vs spinal cord gliomas in adults.
Collapse
Affiliation(s)
- Adriana-Natalia Murgoci
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Marie Duhamel
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Antonella Raffo-Romero
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Khalil Mallah
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Soulaimane Aboulouard
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Christophe Lefebvre
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Firas Kobeissy
- Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Isabelle Fournier
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| | - Monika Zilkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Denisa Maderova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Milan Cizek
- Department of Epizootiology and Parasitology, University of Veterinary Medicine and Pharmacy in Košice, KošiceSlovakia
| | - Dasa Cizkova
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Michel Salzet
- Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, Université Lille, Villeneuve d’Ascq, France
| |
Collapse
|
243
|
Immune cell regulation of glia during CNS injury and disease. Nat Rev Neurosci 2020; 21:139-152. [PMID: 32042145 DOI: 10.1038/s41583-020-0263-9] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
|
244
|
Hashemiaghdam A, Mroczek M. Microglia heterogeneity and neurodegeneration: The emerging paradigm of the role of immunity in Alzheimer's disease. J Neuroimmunol 2020; 341:577185. [PMID: 32045774 DOI: 10.1016/j.jneuroim.2020.577185] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 02/01/2020] [Accepted: 02/01/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is the most common dementia type affecting nearly 44 million people worldwide. Recent findings point to microglia as a significant contributor to neural development, neuroinflammation, and degeneration. Dysregulated immunoactivity in AD has been broadly studied, and current research on animal models enabled us to identify a new cluster of microglia (disease-associated microglia) alongside previously detected glial populations (e.g., plaque-associated microglia, dark microglia, Human Alzheimer's microglia) associated with neuroinflammation and with macrophagic activity. These distinct populations of glia show a spatial distribution within plaques with unique imaging features and distinct gene expression profile. Novel genetic approaches using single-nuclei RNA sequencing (sn-RNA seq) allowed researchers to identify gene expression profiles from fixed human samples. Recent studies, exposing transcriptomic clusters of disease-related cells and analyzing sequenced RNA from sorted myeloid cells, seem to confirm the hypothesis of the central role of glia in the pathogenesis of Alzheimer's disease. These discoveries may shed light on the effects of microglial activation and differences in gene expression profiles, furthering research towards the development of a cell-specific therapy. In this review, we examine recent studies that guide us towards recognizing the role of diverse populations of glial cells and their possible heterogeneous functional states in the pathogenesis of AD in humans.
Collapse
Affiliation(s)
| | - Magdalena Mroczek
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
245
|
Tan YL, Yuan Y, Tian L. Microglial regional heterogeneity and its role in the brain. Mol Psychiatry 2020; 25:351-367. [PMID: 31772305 PMCID: PMC6974435 DOI: 10.1038/s41380-019-0609-8] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023]
Abstract
Microglia have been recently shown to manifest a very interesting phenotypical heterogeneity across different regions in the mammalian central nervous system (CNS). However, the underlying mechanism and functional meaning of this phenomenon are currently unclear. Baseline diversities of adult microglia in their cell number, cellular and subcellular structures, molecular signature as well as relevant functions have been discovered. But recent transcriptomic studies using bulk RNAseq and single-cell RNAseq have produced conflicting results on region-specific signatures of microglia. It is highly speculative whether such spatial heterogeneity contributes to varying sensitivities of individual microglia to the same physiological and pathological signals in different CNS regions, and hence underlie their functional relevance for CNS disease development. This review aims to thoroughly summarize up-to-date knowledge on this specific topic and provide some insights on the potential underlying mechanisms, starting from microgliogenesis. Understanding regional heterogeneity of microglia in the context of their diverse neighboring neurons and other glia may provide an important clue for future development of innovative therapies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yun-Long Tan
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Yi Yuan
- Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Li Tian
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China.
- Institute of Biomedicine and Translational Medicine, Department of Physiology, Faculty of Medicine, University of Tartu, Tartu, Estonia.
| |
Collapse
|
246
|
Prefrontal cortex alterations in glia gene expression in schizophrenia with and without suicide. J Psychiatr Res 2020; 121:31-38. [PMID: 31739114 DOI: 10.1016/j.jpsychires.2019.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/01/2019] [Accepted: 11/08/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Patients with schizophrenia (SCZ) run a lifelong risk of suicide. Alterations in glia activities in the prefrontal cortex (PFC) have been reported in relation to suicide in patients with SCZ. While immune processes in the CNS have been related to the susceptibility and course of SCZ, there are hardly any direct comparisons between individuals with SCZ, both those who died of natural causes and those that committed suicide, and healthy controls. MATERIALS AND METHODS We compared mRNA expression using real time qPCR of 16 glia-related genes in the dorsal lateral prefrontal cortex (DLPFC) and the anterior cingulate cortex (ACC) between 35 patients with SCZ (7 suicide completers and 28 patients who died of natural causes) and 34 well-matched controls without psychiatric or neurological diseases. RESULTS We found an increased expression of the astrocytic gene aldehyde dehydrogenase-1 family member L1 (ALDH1L1) mRNA, a marker involved in dopaminergic activity, in SCZ versus controls. Excluding individuals with SCZ that committed suicide resulted in an elevated expression in the DLPFC of both ALDH1L1 and glutamine synthetase (GS) genes in patients with SCZ, compared to suicide completers and non-psychiatric controls. Regarding microglia genes: in the ACC, homeostatic markers such as chemokine (C-X3-C motif) ligand 1 (CX3CR1) mRNA expression was increased in SCZ without suicide as compared to suicide completers, while no change was found when compared to controls. Another, purinergic receptor 12 (P2RY12) mRNA was exclusively elevated in the ACC of suicide completers, compared to either other group. Triggering receptor expressed on myeloid cells 2 (TREM2) expression, which maintains microglial metabolism, was reduced in non-suicide patients with SCZ, compared to suicide victims and control subjects. CONCLUSIONS Differential changes are found in astrocyte and microglia genes in the PFC subregions in relation to SCZ and suicide, indicating possible disturbances of glia homeostasis in these conditions.
Collapse
|
247
|
Abstract
Microglia are resident macrophages of the CNS that are involved in its development, homeostasis and response to infection and damage. Microglial activation is a common feature of neurological disorders, and although in some instances this activation can be damaging, protective and regenerative functions of microglia have been revealed. The most prominent example of the regenerative functions is a role for microglia in supporting regeneration of myelin after injury, a process that is critical for axonal health and relevant to numerous disorders in which loss of myelin integrity is a prevalent feature, such as multiple sclerosis, Alzheimer disease and motor neuron disease. Although drugs that are intended to promote remyelination are entering clinical trials, the mechanisms by which remyelination is controlled and how microglia are involved are not completely understood. In this Review, we discuss work that has identified novel regulators of microglial activation - including molecular drivers, population heterogeneity and turnover - that might influence their pro-remyelination capacity. We also discuss therapeutic targeting of microglia as a potential approach to promoting remyelination.
Collapse
|
248
|
Hirbec H, Déglon N, Foo LC, Goshen I, Grutzendler J, Hangen E, Kreisel T, Linck N, Muffat J, Regio S, Rion S, Escartin C. Emerging technologies to study glial cells. Glia 2020; 68:1692-1728. [PMID: 31958188 DOI: 10.1002/glia.23780] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Development, physiological functions, and pathologies of the brain depend on tight interactions between neurons and different types of glial cells, such as astrocytes, microglia, oligodendrocytes, and oligodendrocyte precursor cells. Assessing the relative contribution of different glial cell types is required for the full understanding of brain function and dysfunction. Over the recent years, several technological breakthroughs were achieved, allowing "glio-scientists" to address new challenging biological questions. These technical developments make it possible to study the roles of specific cell types with medium or high-content workflows and perform fine analysis of their mutual interactions in a preserved environment. This review illustrates the potency of several cutting-edge experimental approaches (advanced cell cultures, induced pluripotent stem cell (iPSC)-derived human glial cells, viral vectors, in situ glia imaging, opto- and chemogenetic approaches, and high-content molecular analysis) to unravel the role of glial cells in specific brain functions or diseases. It also illustrates the translation of some techniques to the clinics, to monitor glial cells in patients, through specific brain imaging methods. The advantages, pitfalls, and future developments are discussed for each technique, and selected examples are provided to illustrate how specific "gliobiological" questions can now be tackled.
Collapse
Affiliation(s)
- Hélène Hirbec
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nicole Déglon
- Laboratory of Neurotherapies and Neuromodulation, Department of Clinical Neuroscience, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Lynette C Foo
- Neuroimmunology and Neurodegeneration Section, The Neuroscience and Rare Diseases Discovery and Translational Area, F. Hoffman-La Roche, Basel, Switzerland
| | - Inbal Goshen
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jaime Grutzendler
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emilie Hangen
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Neurodegenerative Diseases Laboratory, Université Paris-Sud, Université Paris-Saclay, UMR 9199, Fontenay-aux-Roses, France
| | - Tirzah Kreisel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nathalie Linck
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julien Muffat
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, and Department of Molecular Genetics, The University of Toronto, Toronto, Canada
| | - Sara Regio
- Laboratory of Neurotherapies and Neuromodulation, Department of Clinical Neuroscience, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sybille Rion
- Neuroimmunology and Neurodegeneration Section, The Neuroscience and Rare Diseases Discovery and Translational Area, F. Hoffman-La Roche, Basel, Switzerland
| | - Carole Escartin
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Neurodegenerative Diseases Laboratory, Université Paris-Sud, Université Paris-Saclay, UMR 9199, Fontenay-aux-Roses, France
| |
Collapse
|
249
|
Brioschi S, Zhou Y, Colonna M. Brain Parenchymal and Extraparenchymal Macrophages in Development, Homeostasis, and Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:294-305. [PMID: 31907272 PMCID: PMC7034672 DOI: 10.4049/jimmunol.1900821] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/27/2019] [Indexed: 12/23/2022]
Abstract
Microglia are parenchymal macrophages of the CNS; as professional phagocytes they are important for maintenance of the brain's physiology. These cells are generated through primitive hematopoiesis in the yolk sac and migrate into the brain rudiment after establishment of embryonic circulation. Thereafter, microglia develop in a stepwise fashion, reaching complete maturity after birth. In the CNS, microglia self-renew without input from blood monocytes. Recent RNA-sequencing studies have defined a molecular signature for microglia under homeostasis. However, during disease, microglia undergo remarkable phenotypic changes, which reflect the acquisition of specialized functions tailored to the pathological context. In addition to microglia, the brain-border regions host populations of extraparenchymal macrophages with disparate origins and phenotypes that have recently been delineated. In this review we outline recent findings that provide a deeper understanding of both parenchymal microglia and extraparenchymal brain macrophages in homeostasis and during disease.
Collapse
Affiliation(s)
- Simone Brioschi
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
250
|
Abstract
Microglia are the resident immune cells and professional phagocytes of the central nervous system. However, little is known about the contribution of their phagocytic signaling to the neuropathology and pathophysiology of epilepsy. Here, we summarize and discuss the implications of recent evidence supporting that aberrant microglia phagocytic activity and alterations in phagocytosis signaling molecules occur in association with microglia-neuronal contacts, neuronal/synaptic loss, and spontaneous recurrent seizures in human and preclinical models of epilepsy. This body of evidence provides strong support that the microglial contribution to epileptogenic networks goes beyond inflammation, and suggests that phagocytic signaling molecules may be novel therapeutic targets for epilepsy.
Collapse
Affiliation(s)
| | - Amy L. Brewster
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, USA
| |
Collapse
|