251
|
Sadhukhan T, Bagh MB, Appu AP, Mondal A, Zhang W, Liu A, Mukherjee AB. In a mouse model of INCL reduced S-palmitoylation of cytosolic thioesterase APT1 contributes to microglia proliferation and neuroinflammation. J Inherit Metab Dis 2021; 44:1051-1069. [PMID: 33739454 DOI: 10.1002/jimd.12379] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 01/19/2023]
Abstract
S-palmitoylation is a reversible posttranslational modification in which a 16-carbon saturated fatty acid (generally palmitate) is attached to specific cysteine residues in polypeptides via thioester linkage. Dynamic S-palmitoylation (palmitoylation-depalmitoylation), like phosphorylation-dephosphorylation, regulates the function of numerous proteins, especially in the brain. While a family of 23 palmitoyl-acyl transferases (PATS), commonly known as ZDHHCs, catalyze S-palmitoylation of proteins, the thioesterases, localized either in the cytoplasm (eg, APT1) or in the lysosome (eg, PPT1) mediate depalmitoylation. Previously, we reported that APT1 requires dynamic S-palmitoylation for shuttling between the cytosol and the plasma membrane. APT1 depalmitoylated H-Ras to regulate its signaling pathway that stimulates cell proliferation. Although we demonstrated that APT1 catalyzed its own depalmitoylation, the ZDHHC(s) that S-palmitoylated APT1 had remained unidentified. We report here that ZDHHC5 and ZDHHC23 catalyze APT1 S-palmitoylation. Intriguingly, lysosomal Ppt1-deficiency in Cln1-/- mouse, a reliable animal model of INCL, markedly reduced ZDHHC5 and ZDHHC23 levels. Remarkably, in the brain of these mice decreased ZDHHC5 and ZDHHC23 levels suppressed membrane-bound APT1, thereby, increasing plasma membrane-localized H-Ras, which activated its signaling pathway stimulating microglia proliferation. Increased inflammatory cytokines produced by microglia together with increased complement C1q level contributed to the transformation of astrocytes to neurotoxic A1 phenotype. Importantly, neuroinflammation was ameliorated by treatment of Cln1-/- mice with a PPT1-mimetic small molecule, N-tert(Butyl)hydroxylamine (NtBuHA). Our results revealed a novel pathway to neuropathology in an INCL mouse model and uncovered a previously unrecognized mechanism of the neuroprotective actions of NtBuHA and its potential as a drug target.
Collapse
Affiliation(s)
- Tamal Sadhukhan
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, USA
| | - Maria B Bagh
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, USA
| | - Abhilash P Appu
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, USA
| | - Avisek Mondal
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, USA
| | - Wei Zhang
- Biostatistics and Bioinformatics Branch (HNT72), Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Aiyi Liu
- Biostatistics and Bioinformatics Branch (HNT72), Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Anil B Mukherjee
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
252
|
Alexaki VI. The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives. Cells 2021; 10:cells10071584. [PMID: 34201844 PMCID: PMC8307603 DOI: 10.3390/cells10071584] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Increased life expectancy in combination with modern life style and high prevalence of obesity are important risk factors for development of neurodegenerative diseases. Neuroinflammation is a feature of neurodegenerative diseases, and microglia, the innate immune cells of the brain, are central players in it. The present review discusses the effects of obesity, chronic peripheral inflammation and obesity-associated metabolic and endocrine perturbations, including insulin resistance, dyslipidemia and increased glucocorticoid levels, on microglial function.
Collapse
Affiliation(s)
- Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| |
Collapse
|
253
|
Neckles VN, Feliciano DM. From seed to flower: blossoming of microglia in development and brain repair. Cell Tissue Res 2021; 387:377-389. [PMID: 34151391 DOI: 10.1007/s00441-021-03486-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/09/2021] [Indexed: 12/23/2022]
Abstract
Physiological functions require coordination of processes between diverse organs, tissues, and cells. This integrative view of science has reemerged complementary to the reductionist philosophy of studying individual cell types. An integrative approach has proven particularly powerful within the field of neuroscience where, intermingled among the most numerous neural cell types of the brain, are immune cells called microglia. Microglia act as a line of defense in the CNS by phagocytizing harmful pathogens and cellular debris and by releasing a variety of factors that mediate immune responses. However, microglia are also appreciated as critical mediators of neurophysiology making them a desired target to rectify neuropathological states. The goal of this review is to discuss microglia ontogenesis, referred to as microgliogenesis, a term that encompasses the events that drive the production, differentiation, migration, and maturation of microglia and opportunities to target microglia for brain repair.
Collapse
Affiliation(s)
- Victoria N Neckles
- Department of Biological Sciences, Clemson University, Clemson, SC, 29634-0314, USA
| | - David M Feliciano
- Department of Biological Sciences, Clemson University, Clemson, SC, 29634-0314, USA.
| |
Collapse
|
254
|
Woolf Z, Stevenson TJ, Lee K, Jung Y, Park TI, Curtis MA, Montgomery JM, Dragunow M. Isolation of adult mouse microglia using their in vitro adherent properties. STAR Protoc 2021; 2:100518. [PMID: 34027479 PMCID: PMC8121984 DOI: 10.1016/j.xpro.2021.100518] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Microglia are the primary innate immune effectors of the central nervous system. Although numerous protocols have been developed to isolate fetal mouse microglia, the isolation of adult mouse microglia has proven more difficult. Here, we present a simple, widely accessible protocol to isolate pure microglia cultures from 4- to 14-month-old mouse brains using their adherent properties in vitro. These isolated microglia recapitulate the adherent properties of adult human microglia and present a more suitable model for studying age-related diseases. For complete details on the use and execution of this protocol in adult human microglia, please refer to Rustenhoven et al. (2016).
Collapse
Affiliation(s)
- Zoe Woolf
- Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, Private Bag 92019, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, Private Bag 92019, New Zealand
| | - Taylor J. Stevenson
- Centre for Brain Research, The University of Auckland, Auckland, Private Bag 92019, New Zealand
- Department of Anatomy and Medical Imaging, The University of Auckland, Auckland, Private Bag 92019, New Zealand
| | - Kevin Lee
- Centre for Brain Research, The University of Auckland, Auckland, Private Bag 92019, New Zealand
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, Private Bag 92019, New Zealand
| | - Yewon Jung
- Centre for Brain Research, The University of Auckland, Auckland, Private Bag 92019, New Zealand
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, Private Bag 92019, New Zealand
| | - Thomas I.H. Park
- Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, Private Bag 92019, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, Private Bag 92019, New Zealand
| | - Maurice A. Curtis
- Centre for Brain Research, The University of Auckland, Auckland, Private Bag 92019, New Zealand
- Department of Anatomy and Medical Imaging, The University of Auckland, Auckland, Private Bag 92019, New Zealand
| | - Johanna M. Montgomery
- Centre for Brain Research, The University of Auckland, Auckland, Private Bag 92019, New Zealand
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, Private Bag 92019, New Zealand
| | - Michael Dragunow
- Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, Private Bag 92019, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, Private Bag 92019, New Zealand
| |
Collapse
|
255
|
Hammond BP, Manek R, Kerr BJ, Macauley MS, Plemel JR. Regulation of microglia population dynamics throughout development, health, and disease. Glia 2021; 69:2771-2797. [PMID: 34115410 DOI: 10.1002/glia.24047] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/20/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022]
Abstract
The dynamic expansions and contractions of the microglia population in the central nervous system (CNS) to achieve homeostasis are likely vital for their function. Microglia respond to injury or disease but also help guide neurodevelopment, modulate neural circuitry throughout life, and direct regeneration. Throughout these processes, microglia density changes, as does the volume of area that each microglia surveys. Given that microglia are responsible for sensing subtle alterations to their environment, a change in their density could affect their capacity to mobilize rapidly. In this review, we attempt to synthesize the current literature on the ligands and conditions that promote microglial proliferation across development, adulthood, and neurodegenerative conditions. Microglia display an impressive proliferative capacity during development and in neurodegenerative diseases that is almost completely absent at homeostasis. However, the appropriate function of microglia in each state is critically dependent on density fluctuations that are primarily induced by proliferation. Proliferation is a natural microglial response to insult and often serves neuroprotective functions. In contrast, inappropriate microglial proliferation, whether too much or too little, often precipitates undesirable consequences for nervous system health. Thus, fluctuations in the microglia population are tightly regulated to ensure these immune cells can execute their diverse functions.
Collapse
Affiliation(s)
- Brady P Hammond
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Rupali Manek
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
256
|
Aires V, Coulon-Bainier C, Pavlovic A, Ebeling M, Schmucki R, Schweitzer C, Kueng E, Gutbier S, Harde E. CD22 Blockage Restores Age-Related Impairments of Microglia Surveillance Capacity. Front Immunol 2021; 12:684430. [PMID: 34140954 PMCID: PMC8204252 DOI: 10.3389/fimmu.2021.684430] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/17/2021] [Indexed: 11/13/2022] Open
Abstract
Microglia, the innate immune cells of the brain, are essential for maintaining homeostasis by their ramified, highly motile processes and for orchestrating the immune response to pathological stimuli. They are implicated in several neurodegenerative diseases like Alzheimer's and Parkinson's disease. One commonality of these diseases is their strong correlation with aging as the highest risk factor and studying age-related alterations in microglia physiology and associated signaling mechanism is indispensable for a better understanding of age-related pathomechanisms. CD22 has been identified as a modifier of microglia phagocytosis in a recent study, but not much is known about the function of CD22 in microglia. Here we show that CD22 surface levels are upregulated in aged versus adult microglia. Furthermore, in the amyloid mouse model PS2APP, Aβ-containing microglia also exhibit increased CD22 signal. To assess the impact of CD22 blockage on microglia morphology and dynamics, we have established a protocol to image microglia process motility in acutely prepared brain slices from CX3CR1-GFP reporter mice. We observed a significant reduction of microglial ramification and surveillance capacity in brain slices from aged versus adult mice. The age-related decrease in surveillance can be restored by antibody-mediated CD22 blockage in aged mice, whereas surveillance in adult mice is not affected by CD22 inhibition. Moreover to complement the results obtained in mice, we show that human iPSC-derived macrophages exhibit an increased phagocytic capacity upon CD22 blockage. Downstream analysis of antibody-mediated CD22 inhibition revealed an influence on BMP and TGFβ associated gene networks. Our results demonstrate CD22 as a broad age-associated modulator of microglia functionality with potential implications for neurodegenerative disorders.
Collapse
Affiliation(s)
- Vanessa Aires
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.,Department of Neurology, Medical Center - University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Claire Coulon-Bainier
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Anto Pavlovic
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Martin Ebeling
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Roland Schmucki
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Christophe Schweitzer
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Erich Kueng
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Simon Gutbier
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Eva Harde
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
257
|
Chen W, Xie L, Yu F, Li Y, Chen C, Xie W, Huang T, Zhang Y, Zhang S, Li P. Zebrafish as a Model for In-Depth Mechanistic Study for Stroke. Transl Stroke Res 2021; 12:695-710. [PMID: 34050491 DOI: 10.1007/s12975-021-00907-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022]
Abstract
Stroke is one of the world's leading causes of death and disability, posing enormous burden to the society. However, the pathogenesis and mechanisms that underlie brain injury and brain repair remain largely unknown. There's an unmet need of in-depth mechanistic research in this field. Zebrafish (Danio rerio) is a powerful tool in brain science research mainly due to its small size and transparent body, high genome synteny with human, and similar nervous system structures. It can be used to establish both hemorrhagic and ischemic stroke models easily and effectively through different ways. After the establishment of stroke model, research methods including behavioral test, in vivo imaging, and drug screening are available to explore mechanisms that underlie the brain injury and brain repair after stroke. This review focuses on the advantages and the feasibility of zebrafish stroke model, and will also introduce the key methods available for stroke studies in zebrafish, which may drive future mechanistic studies in the pursuit of discovering novel therapeutic targets for stroke patients.
Collapse
Affiliation(s)
- Weijie Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Lv Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Fang Yu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yan Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Chen Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Wanqing Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Tingting Huang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Song Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| |
Collapse
|
258
|
Zilberman A, Cornelison RC. Microphysiological models of the central nervous system with fluid flow. Brain Res Bull 2021; 174:72-83. [PMID: 34029679 DOI: 10.1016/j.brainresbull.2021.05.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022]
Abstract
There are over 1,000 described neurological and neurodegenerative disorders affecting nearly 100 million Americans - roughly one third of the U.S. population. Collectively, treatment of neurological conditions is estimated to cost $800 billion every year. Lowering this societal burden will require developing better model systems in which to study these diverse disorders. Microphysiological systems are promising tools for modeling healthy and diseased neural tissues to study mechanisms and treatment of neuropathology. One major benefit of microphysiological systems is the ability to incorporate biophysical forces, namely the forces derived from biological fluid flow. Fluid flow in the central nervous system (CNS) is a complex but important element of physiology, and pathologies as diverse as traumatic or ischemic injury, cancer, neurodegenerative disease, and natural aging have all been found to alter flow pathways. In this review, we summarize recent advances in three-dimensional microphysiological systems for studying the biology and therapy of CNS disorders and highlight the ability and growing need to incorporate biological fluid flow in these miniaturized model systems.
Collapse
Affiliation(s)
- Aleeza Zilberman
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, United States
| | - R Chase Cornelison
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, United States.
| |
Collapse
|
259
|
Transcriptional signature in microglia associated with Aβ plaque phagocytosis. Nat Commun 2021; 12:3015. [PMID: 34021136 PMCID: PMC8140091 DOI: 10.1038/s41467-021-23111-1] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 04/13/2021] [Indexed: 12/13/2022] Open
Abstract
The role of microglia cells in Alzheimer’s disease (AD) is well recognized, however their molecular and functional diversity remain unclear. Here, we isolated amyloid plaque-containing (using labelling with methoxy-XO4, XO4+) and non-containing (XO4−) microglia from an AD mouse model. Transcriptomics analysis identified different transcriptional trajectories in ageing and AD mice. XO4+ microglial transcriptomes demonstrated dysregulated expression of genes associated with late onset AD. We further showed that the transcriptional program associated with XO4+ microglia from mice is present in a subset of human microglia isolated from brains of individuals with AD. XO4− microglia displayed transcriptional signatures associated with accelerated ageing and contained more intracellular post-synaptic material than XO4+ microglia, despite reduced active synaptosome phagocytosis. We identified HIF1α as potentially regulating synaptosome phagocytosis in vitro using primary human microglia, and BV2 mouse microglial cells. Together, these findings provide insight into molecular mechanisms underpinning the functional diversity of microglia in AD. Microglia associated with Aβ plaques may have a distinct transcriptional signature compared to those in plaque-free areas of the brain in Alzheimer’s disease (AD) models. Here the authors show that amyloid plaque phagocytosis is associated with a specific microglia transcriptional signature in a mouse model of AD.
Collapse
|
260
|
Zago G, Saavedra PHV, Keshari KR, Perry JSA. Immunometabolism of Tissue-Resident Macrophages - An Appraisal of the Current Knowledge and Cutting-Edge Methods and Technologies. Front Immunol 2021; 12:665782. [PMID: 34025667 PMCID: PMC8138590 DOI: 10.3389/fimmu.2021.665782] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/07/2021] [Indexed: 12/23/2022] Open
Abstract
Tissue-resident macrophages exist in unique environments, or niches, that inform their identity and function. There is an emerging body of literature suggesting that the qualities of this environment, such as the types of cells and debris they eat, the intercellular interactions they form, and the length of time spent in residence, collectively what we call habitare, directly inform their metabolic state. In turn, a tissue-resident macrophage’s metabolic state can inform their function, including whether they resolve inflammation and protect the host from excessive perturbations of homeostasis. In this review, we summarize recent work that seeks to understand the metabolic requirements for tissue-resident macrophage identity and maintenance, for how they respond to inflammatory challenges, and for how they perform homeostatic functions or resolve inflammatory insults. We end with a discussion of the emerging technologies that are enabling, or will enable, in situ study of tissue-resident macrophage metabolism.
Collapse
Affiliation(s)
- Giulia Zago
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Pedro H V Saavedra
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Kayvan R Keshari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Justin S A Perry
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
261
|
Si Z, Wang X. Stem Cell Therapies in Alzheimer's Disease: Applications for Disease Modeling. J Pharmacol Exp Ther 2021; 377:207-217. [PMID: 33558427 DOI: 10.1124/jpet.120.000324] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/03/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with complex pathologic and biologic characteristics. Extracellular β-amyloid deposits, such as senile plaques, and intracellular aggregation of hyperphosphorylated tau, such as neurofibrillary tangles, remain the main neuropathological criteria for the diagnosis of AD. There is currently no effective treatment of the disease, and many clinical trials have failed to prove any benefits of new therapeutics. More recently, there has been increasing interest in harnessing the potential of stem cell technologies for drug discovery, disease modeling, and cell therapies, which have been used to study an array of human conditions, including AD. The recently developed and optimized induced pluripotent stem cell (iPSC) technology is a critical platform for screening anti-AD drugs and understanding mutations that modify AD. Neural stem cell (NSC) transplantation has been investigated as a new therapeutic approach to treat neurodegenerative diseases. Mesenchymal stem cells (MSCs) also exhibit considerable potential to treat neurodegenerative diseases by secreting growth factors and exosomes, attenuating neuroinflammation. This review highlights recent progress in stem cell research and the translational applications and challenges of iPSCs, NSCs, and MSCs as treatment strategies for AD. Even though these treatments are still in relative infancy, these developing stem cell technologies hold considerable promise to combat AD and other neurodegenerative disorders. SIGNIFICANCE STATEMENT: Alzheimer's disease (AD) is a neurodegenerative disease that results in learning and memory defects. Although some drugs have been approved for AD treatment, fewer than 20% of patients with AD benefit from these drugs. Therapies based on stem cells, including induced pluripotent stem cells, neural stem cells, and mesenchymal stem cells, provide promising therapeutic strategies for AD.
Collapse
Affiliation(s)
- Zizhen Si
- Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
| | - Xidi Wang
- Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
| |
Collapse
|
262
|
Abstract
Tissue-resident macrophages are present in most tissues with developmental, self-renewal, or functional attributes that do not easily fit into a textbook picture of a plastic and multifunctional macrophage originating from hematopoietic stem cells; nor does it fit a pro- versus anti-inflammatory paradigm. This review presents and discusses current knowledge on the developmental biology of macrophages from an evolutionary perspective focused on the function of macrophages, which may aid in study of developmental, inflammatory, tumoral, and degenerative diseases. We also propose a framework to investigate the functions of macrophages in vivo and discuss how inherited germline and somatic mutations may contribute to the roles of macrophages in diseases.
Collapse
Affiliation(s)
- Nehemiah Cox
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Maria Pokrovskii
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| |
Collapse
|
263
|
Minhas PS, Durairaj AS, Andreasson KI. The Dueling Duo: IL10 and TNF Face Off in Microglial Recovery from Endotoxin Challenge. Immunity 2021; 53:897-899. [PMID: 33207212 DOI: 10.1016/j.immuni.2020.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The molecular mechanisms that restore microglial quiescence after acute stimulation remain largely unexplored, unlike those that drive microglial activation. In this issue of Immunity, Shemer et al. discover that the microglial IL-10 receptor counteracts the pro-inflammatory effects of TNF to allow restoration of microglial quiescence after peripheral endotoxin challenge.
Collapse
Affiliation(s)
| | | | - Katrin I Andreasson
- Department of Neurology, Stanford University, Stanford, CA, USA; Wu-Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Program in Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
264
|
Colombo AV, Sadler RK, Llovera G, Singh V, Roth S, Heindl S, Sebastian Monasor L, Verhoeven A, Peters F, Parhizkar S, Kamp F, Gomez de Aguero M, MacPherson AJ, Winkler E, Herms J, Benakis C, Dichgans M, Steiner H, Giera M, Haass C, Tahirovic S, Liesz A. Microbiota-derived short chain fatty acids modulate microglia and promote Aβ plaque deposition. eLife 2021; 10:e59826. [PMID: 33845942 PMCID: PMC8043748 DOI: 10.7554/elife.59826] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 03/28/2021] [Indexed: 12/12/2022] Open
Abstract
Previous studies have identified a crucial role of the gut microbiome in modifying Alzheimer's disease (AD) progression. However, the mechanisms of microbiome-brain interaction in AD were so far unknown. Here, we identify microbiota-derived short chain fatty acids (SCFA) as microbial metabolites which promote Aβ deposition. Germ-free (GF) AD mice exhibit a substantially reduced Aβ plaque load and markedly reduced SCFA plasma concentrations; conversely, SCFA supplementation to GF AD mice increased the Aβ plaque load to levels of conventionally colonized (specific pathogen-free [SPF]) animals and SCFA supplementation to SPF mice even further exacerbated plaque load. This was accompanied by the pronounced alterations in microglial transcriptomic profile, including upregulation of ApoE. Despite increased microglial recruitment to Aβ plaques upon SCFA supplementation, microglia contained less intracellular Aβ. Taken together, our results demonstrate that microbiota-derived SCFA are critical mediators along the gut-brain axis which promote Aβ deposition likely via modulation of the microglial phenotype.
Collapse
Affiliation(s)
| | - Rebecca Katie Sadler
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU MunichMunichGermany
| | - Gemma Llovera
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU MunichMunichGermany
| | - Vikramjeet Singh
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU MunichMunichGermany
| | - Stefan Roth
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU MunichMunichGermany
| | - Steffanie Heindl
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU MunichMunichGermany
| | | | - Aswin Verhoeven
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC)LeidenNetherlands
| | - Finn Peters
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| | - Samira Parhizkar
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Frits Kamp
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Mercedes Gomez de Aguero
- Maurice Müller Laboratories (DKF), Universitätsklinik für Viszerale Chirurgie und Medizin InselspitalBernSwitzerland
| | - Andrew J MacPherson
- Maurice Müller Laboratories (DKF), Universitätsklinik für Viszerale Chirurgie und Medizin InselspitalBernSwitzerland
| | - Edith Winkler
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University MunichMunichGermany
| | - Corinne Benakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU MunichMunichGermany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Harald Steiner
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC)LeidenNetherlands
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität MünchenMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| |
Collapse
|
265
|
Chang CF, Goods BA, Askenase MH, Beatty HE, Osherov A, DeLong JH, Hammond MD, Massey J, Landreneau M, Love JC, Sansing LH. Divergent Functions of Tissue-Resident and Blood-Derived Macrophages in the Hemorrhagic Brain. Stroke 2021; 52:1798-1808. [PMID: 33840225 DOI: 10.1161/strokeaha.120.032196] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Che-Feng Chang
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei (C.-F.C.).,Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Brittany A Goods
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge (B.A.G., J.C.L.)
| | - Michael H Askenase
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT.,Immunobiology (M.H.A., H.E.B., J.H.D., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Hannah E Beatty
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT.,Immunobiology (M.H.A., H.E.B., J.H.D., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Artem Osherov
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Jonathan H DeLong
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT.,Immunobiology (M.H.A., H.E.B., J.H.D., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Matthew D Hammond
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Jordan Massey
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - Margaret Landreneau
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT
| | - J Christopher Love
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge (B.A.G., J.C.L.)
| | - Lauren H Sansing
- Departments of Neurology (C.-F.C., M.H.A., H.E.B., A.O., J.H.D., M.D.H., J.M., M.L., L.H.S.), Yale University School of Medicine, New Haven, CT.,Immunobiology (M.H.A., H.E.B., J.H.D., L.H.S.), Yale University School of Medicine, New Haven, CT
| |
Collapse
|
266
|
Chen Y, Hong T, Chen F, Sun Y, Wang Y, Cui L. Interplay Between Microglia and Alzheimer's Disease-Focus on the Most Relevant Risks: APOE Genotype, Sex and Age. Front Aging Neurosci 2021; 13:631827. [PMID: 33897406 PMCID: PMC8060487 DOI: 10.3389/fnagi.2021.631827] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 03/18/2021] [Indexed: 12/20/2022] Open
Abstract
As the main immune cells of the central nervous system (CNS), microglia regulates normal development, homeostasis and general brain physiology. These functions put microglia at the forefront of CNS repair and recovery. Uncontrolled activation of microglia is related to the course of neurodegenerative diseases such as Alzheimer’s disease. It is clear that the classic pathologies of amyloid β (Aβ) and Tau are usually accompanied by the activation of microglia, and the activation of microglia also serves as an early event in the pathogenesis of AD. Therefore, during the occurrence and development of AD, the key susceptibility factors for AD—apolipoprotein E (APOE) genotype, sex and age—may further interact with microglia to exacerbate neurodegeneration. In this review, we discuss the role of microglia in the progression of AD related to the three risk factors for AD: APOE genotype, sex and aging. APOE-expressing microglia accumulates around Aβ plaques, and the presence of APOE4 may disrupt the phagocytosis of Aβ aggregates and aggravate neurodegeneration in Tau disease models. In addition, females have a high incidence of AD, and normal female microglia and estrogen have protective effects under normal conditions. However, under the influence of AD, female microglia seem to lose their protective effect and instead accelerate the course of AD. Aging, another major risk factor, may increase the sensitivity of microglia, leading to the exacerbation of microglial dysfunction in elderly AD. Obviously, in the role of microglia in AD, the three main risk factors of APOE, sex, and aging are not independent and have synergistic effects that contribute to the risk of AD. Moreover, new microglia can replace dysfunctional microglia after microglial depletion, which is a new promising strategy for AD treatment.
Collapse
Affiliation(s)
- Yanting Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Tingting Hong
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Feng Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yuanhong Sun
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Yan Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
267
|
Jesudasan SJB, Gupta SJ, Churchward MA, Todd KG, Winship IR. Inflammatory Cytokine Profile and Plasticity of Brain and Spinal Microglia in Response to ATP and Glutamate. Front Cell Neurosci 2021; 15:634020. [PMID: 33889075 PMCID: PMC8057348 DOI: 10.3389/fncel.2021.634020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/10/2021] [Indexed: 11/27/2022] Open
Abstract
Microglia are the primary cells in the central nervous system that identify and respond to injury or damage. Such a perturbation in the nervous system induces the release of molecules including ATP and glutamate that act as damage-associated molecular patterns (DAMPs). DAMPs are detected by microglia, which then regulate the inflammatory response in a manner sensitive to their surrounding environment. The available data indicates that ATP and glutamate can induce the release of pro inflammatory factors TNF (tumor necrosis factor), IL-1β (interleukin 1 beta), and NO (nitric oxide) from microglia. However, non-physiological concentrations of ATP and glutamate were often used to derive these insights. Here, we have compared the response of spinal cord microglia (SM) relative to brain microglia (BM) using physiologically relevant concentrations of glutamate and ATP that mimic injured conditions in the central nervous system. The data show that ATP and glutamate are not significant modulators of the release of cytokines from either BM or SM. Consistent with previous studies, spinal microglia exhibited a general trend toward reduced release of inflammatory cytokines relative to brain-derived microglia. Moreover, we demonstrate that the responses of microglia to these DAMPs can be altered by modifying the biochemical milieu in their surrounding environment. Preconditioning brain derived microglia with media from spinal cord derived mixed glial cultures shifted their release of IL-1ß and IL-6 to a less inflammatory phenotype consistent with spinal microglia.
Collapse
Affiliation(s)
- Sam Joshva Baskar Jesudasan
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Somnath J. Gupta
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Matthew A. Churchward
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Biology and Environmental Sciences, Concordia University of Edmonton, Edmonton, AB, Canada
| | - Kathryn G. Todd
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Ian R. Winship
- Neurochemical Research Unit, Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
268
|
Tavares-Gomes L, Monney C, Neuhaus G, Francisco D, Solis D, Summerfield A, Erny D, Jagannathan V, Oevermann A. Transcriptome of microglia reveals a species-specific expression profile in bovines with conserved and new signature genes. Glia 2021; 69:1932-1949. [PMID: 33811399 DOI: 10.1002/glia.24002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 11/09/2022]
Abstract
Evidence is growing that microglia adopt different roles than monocyte-derived macrophages (MDM) during CNS injury. However, knowledge about their function in the pathogenesis of neuroinfections is only rudimentary. Cattle are frequently affected by neuroinfections that are either zoonotic or related to diseases in humans, and, hence, studies of bovine neuroinfections as a natural disease model may generate fundamental data on their pathogenesis potentially translatable to humans. We investigated the transcriptomic landscape and lineage markers of bovine microglia and MDM. Although bovine microglia expressed most microglial signature genes known from humans and mice, they exhibited a species-specific transcriptomic profile, including strikingly low expression of TMEM119 and enrichment of the two scavenger receptors MEGF10 and LY75. P2RY12 was amongst the most enriched genes in bovine microglia, and antibodies against P2RY12 labeled specifically resting microglia, but also reactive microglia within neuroinfection foci in-situ. On the other hand, F13A1 was amongst the most enriched genes in bovine monocytes and MDM and, additionally, the encoded protein was expressed in-situ in monocytes and MDM in the inflamed brain but not in microglia, making it a promising marker for infiltrating MDM in the brain. In culture, primary bovine microglia downregulated signature genes, expressed markers of activation, and converged their transcriptome to MDM. However, they retained several microglia signature genes that clearly distinguished them from bovine MDM, making them a promising in-vitro tool to study mechanisms of microglia-pathogen interactions.
Collapse
Affiliation(s)
- Leticia Tavares-Gomes
- Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Camille Monney
- Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Géraldine Neuhaus
- Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - David Francisco
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.,Interfaculty of Bioinformatics Unit, University of Bern, Bern, Switzerland.,Swiss Institute of Bioinformatics, Switzerland
| | - Diana Solis
- Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,University of Fribourg, Bern, Switzerland
| | - Artur Summerfield
- Faculty of Veterinary Medicine, Institute of Virology and Immunology, University of Bern, Bern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Daniel Erny
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Anna Oevermann
- Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
269
|
Grassivaro F, Martino G, Farina C. The phenotypic convergence between microglia and peripheral macrophages during development and neuroinflammation paves the way for new therapeutic perspectives. Neural Regen Res 2021; 16:635-637. [PMID: 33063713 PMCID: PMC8067915 DOI: 10.4103/1673-5374.295272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 04/27/2020] [Accepted: 06/13/2020] [Indexed: 12/27/2022] Open
Abstract
Microglia, the tissue resident macrophages of the brain, are increasingly recognized as key players for central nervous system development and homeostasis. They are long-lived cells deriving from a transient wave of yolk-sac derived erythro-myeloid progenitors early in development. Their unique ontology has prompted the search for specific markers to be used for their selective investigation and manipulation. The first generation of genome-wide expression studies has provided a bundle of transcripts (such as Olfml3, Fcrls, Tmem119, P2ry12, Gpr34, and Siglech) useful to distinguish microglia from peripheral macrophages. However, more recent reports have revealed that microglial phenotype is constantly shaped by the microenvironment in a time-, and context-dependent manner. In this article, we review data that provide additional pieces to this complex scenario and show the existence of unexpected phenotypic convergence between microglia and peripheral macrophages at certain developmental stages and under pathological conditions. These observations suggest that the two cell types act synergically boosting their mutual activities depending on the microenvironment. This novel information about the biology of microglia and peripheral macrophages sheds new light about their therapeutic potential for neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Francesca Grassivaro
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Gianvito Martino
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
270
|
van Wageningen TA, Antonovaite N, Paardekam E, Brevé JJP, Iannuzzi D, van Dam AM. Viscoelastic properties of white and gray matter-derived microglia differentiate upon treatment with lipopolysaccharide but not upon treatment with myelin. J Neuroinflammation 2021; 18:83. [PMID: 33781276 PMCID: PMC8008683 DOI: 10.1186/s12974-021-02134-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/18/2021] [Indexed: 01/16/2023] Open
Abstract
Background The biomechanical properties of the brain have increasingly been shown to relate to brain pathology in neurological diseases, including multiple sclerosis (MS). Inflammation and demyelination in MS induce significant changes in brain stiffness which can be linked to the relative abundance of glial cells in lesions. We hypothesize that the biomechanical, in addition to biochemical, properties of white (WM) and gray matter (GM)-derived microglia may contribute to the differential microglial phenotypes as seen in MS WM and GM lesions. Methods Primary glial cultures from WM or GM of rat adult brains were treated with either lipopolysaccharide (LPS), myelin, or myelin+LPS for 24 h or left untreated as a control. After treatment, microglial cells were indented using dynamic indentation to determine the storage and loss moduli reflecting cell elasticity and cell viscosity, respectively, and subsequently fixed for immunocytochemical analysis. In parallel, gene expression of inflammatory-related genes were measured using semi-quantitative RT-PCR. Finally, phagocytosis of myelin was determined as well as F-actin visualized to study the cytoskeletal changes. Results WM-derived microglia were significantly more elastic and more viscous than microglia derived from GM. This heterogeneity in microglia biomechanical properties was also apparent when treated with LPS when WM-derived microglia decreased cell elasticity and viscosity, and GM-derived microglia increased elasticity and viscosity. The increase in elasticity and viscosity observed in GM-derived microglia was accompanied by an increase in Tnfα mRNA and reorganization of F-actin which was absent in WM-derived microglia. In contrast, when treated with myelin, both WM- and GM-derived microglia phagocytose myelin decrease their elasticity and viscosity. Conclusions In demyelinating conditions, when myelin debris is phagocytized, as in MS lesions, it is likely that the observed differences in WM- versus GM-derived microglia biomechanics are mainly due to a difference in response to inflammation, rather than to the event of demyelination itself. Thus, the differential biomechanical properties of WM and GM microglia may add to their differential biochemical properties which depend on inflammation present in WM and GM lesions of MS patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02134-x.
Collapse
Affiliation(s)
- Thecla A van Wageningen
- Department of Anatomy & Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Nelda Antonovaite
- Department of Physics and Astronomy and LaserLaB, VU Amsterdam, Amsterdam, The Netherlands
| | - Erik Paardekam
- Department of Physics and Astronomy and LaserLaB, VU Amsterdam, Amsterdam, The Netherlands
| | - John J P Brevé
- Department of Anatomy & Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Davide Iannuzzi
- Department of Physics and Astronomy and LaserLaB, VU Amsterdam, Amsterdam, The Netherlands
| | - Anne-Marie van Dam
- Department of Anatomy & Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands.
| |
Collapse
|
271
|
Lanjewar SN, Sloan SA. Growing Glia: Cultivating Human Stem Cell Models of Gliogenesis in Health and Disease. Front Cell Dev Biol 2021; 9:649538. [PMID: 33842475 PMCID: PMC8027322 DOI: 10.3389/fcell.2021.649538] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/25/2021] [Indexed: 12/31/2022] Open
Abstract
Glia are present in all organisms with a central nervous system but considerably differ in their diversity, functions, and numbers. Coordinated efforts across many model systems have contributed to our understanding of glial-glial and neuron-glial interactions during nervous system development and disease, but human glia exhibit prominent species-specific attributes. Limited access to primary samples at critical developmental timepoints constrains our ability to assess glial contributions in human tissues. This challenge has been addressed throughout the past decade via advancements in human stem cell differentiation protocols that now offer the ability to model human astrocytes, oligodendrocytes, and microglia. Here, we review the use of novel 2D cell culture protocols, 3D organoid models, and bioengineered systems derived from human stem cells to study human glial development and the role of glia in neurodevelopmental disorders.
Collapse
Affiliation(s)
| | - Steven A. Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
272
|
Baxter PS, Dando O, Emelianova K, He X, McKay S, Hardingham GE, Qiu J. Microglial identity and inflammatory responses are controlled by the combined effects of neurons and astrocytes. Cell Rep 2021; 34:108882. [PMID: 33761343 PMCID: PMC7994374 DOI: 10.1016/j.celrep.2021.108882] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 01/07/2021] [Accepted: 02/25/2021] [Indexed: 12/05/2022] Open
Abstract
Microglia, brain-resident macrophages, require instruction from the CNS microenvironment to maintain their identity and morphology and regulate inflammatory responses, although what mediates this is unclear. Here, we show that neurons and astrocytes cooperate to promote microglial ramification, induce expression of microglial signature genes ordinarily lost in vitro and in age and disease in vivo, and repress infection- and injury-associated gene sets. The influence of neurons and astrocytes separately on microglia is weak, indicative of synergies between these cell types, which exert their effects via a mechanism involving transforming growth factor β2 (TGF-β2) signaling. Neurons and astrocytes also combine to provide immunomodulatory cues, repressing primed microglial responses to weak inflammatory stimuli (without affecting maximal responses) and consequently limiting the feedback effects of inflammation on the neurons and astrocytes themselves. These findings explain why microglia isolated ex vivo undergo de-differentiation and inflammatory deregulation and point to how disease- and age-associated changes may be counteracted.
Collapse
Affiliation(s)
- Paul S Baxter
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4TJ, UK; Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Owen Dando
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4TJ, UK; Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Katie Emelianova
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4TJ, UK; Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Xin He
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4TJ, UK; Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sean McKay
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4TJ, UK; Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Giles E Hardingham
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4TJ, UK; Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK.
| | - Jing Qiu
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, Edinburgh EH16 4TJ, UK; Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK.
| |
Collapse
|
273
|
Jolivel V, Brun S, Binamé F, Benyounes J, Taleb O, Bagnard D, De Sèze J, Patte-Mensah C, Mensah-Nyagan AG. Microglial Cell Morphology and Phagocytic Activity Are Critically Regulated by the Neurosteroid Allopregnanolone: A Possible Role in Neuroprotection. Cells 2021; 10:698. [PMID: 33801063 PMCID: PMC8004004 DOI: 10.3390/cells10030698] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Microglial cells are key players in neural pathogenesis and microglial function regulation appears to be pivotal in controlling neuroinflammatory/neurological diseases. Here, we investigated the effects and mechanism of action of neurosteroid allopregnanolone (ALLO) on murine microglial BV-2 cells and primary microglia in order to determine ALLO-induced immunomodulatory potential and to provide new insights for the development of both natural and safe neuroprotective strategies targeting microglia. Indeed, ALLO-treatment is increasingly suggested as beneficial in various models of neurological disorders but the underlying mechanisms have not been elucidated. Therefore, the microglial cells were cultured with various serum concentrations to mimic the blood-brain-barrier rupture and to induce their activation. Proliferation, viability, RT-qPCR, phagocytosis, and morphology analyzes, as well as migration with time-lapse imaging and quantitative morphodynamic methods, were combined to investigate ALLO actions on microglia. BV-2 cells express subunits of GABA-A receptor that mediates ALLO activity. ALLO (10µM) induced microglial cell process extension and decreased migratory capacity. Interestingly, ALLO modulated the phagocytic activity of BV-2 cells and primary microglia. Our results, which show a direct effect of ALLO on microglial morphology and phagocytic function, suggest that the natural neurosteroid-based approach may contribute to developing effective strategies against neurological disorders that are evoked by microglia-related abnormalities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ayikoe-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 1 rue Eugène Boeckel, 67000 Strasbourg, France; (V.J.); (S.B.); (F.B.); (J.B.); (O.T.); (D.B.); (J.D.S.); (C.P.-M.)
| |
Collapse
|
274
|
Andoh M, Koyama R. Assessing Microglial Dynamics by Live Imaging. Front Immunol 2021; 12:617564. [PMID: 33763064 PMCID: PMC7982483 DOI: 10.3389/fimmu.2021.617564] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Microglia are highly dynamic in the brain in terms of their ability to migrate, proliferate, and phagocytose over the course of an individual's life. Real-time imaging is a useful tool to examine how microglial behavior is regulated and how it affects the surrounding environment. However, microglia are sensitive to environmental stimuli, so they possibly change their state during live imaging in vivo, mainly due to surgical damage, and in vitro due to various effects associated with culture conditions. Therefore, it is difficult to perform live imaging without compromising the properties of the microglia under physiological conditions. To overcome this barrier, various experimental conditions have been developed; recently, it has become possible to perform live imaging of so-called surveillant microglia in vivo, ex vivo, and in vitro, although there are various limitations. Now, we can choose in vivo, ex vivo, or in vitro live imaging systems according to the research objective. In this review, we discuss the advantages and disadvantages of each experimental system and outline the physiological significance and molecular mechanisms of microglial behavior that have been elucidated by live imaging.
Collapse
Affiliation(s)
- Megumi Andoh
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
275
|
Woodburn SC, Bollinger JL, Wohleb ES. Synaptic and behavioral effects of chronic stress are linked to dynamic and sex-specific changes in microglia function and astrocyte dystrophy. Neurobiol Stress 2021; 14:100312. [PMID: 33748354 PMCID: PMC7970222 DOI: 10.1016/j.ynstr.2021.100312] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/28/2021] [Accepted: 02/26/2021] [Indexed: 01/13/2023] Open
Abstract
Emerging evidence indicates that males and females display different neurobiological responses to chronic stress which contribute to varied behavioral adaptations. In particular, pyramidal neurons undergo dendritic atrophy and synapse loss in the prefrontal cortex (PFC) of male, but not female, mice. Our recent work shows that chronic stress also provokes microglia-mediated neuronal remodeling, which contributes to synaptic deficits in the PFC and associated behavioral consequences in males. Separate studies indicate that chronic stress promotes astrocyte dystrophy in the PFC which is associated with behavioral despair. Notably, these prior reports focused primarily on stress effects in males. In the present studies, male and female mice were exposed to 14 or 28 days of chronic unpredictable stress (CUS) to assess molecular and cellular adaptations of microglia, astrocytes, and neurons in the medial PFC. Consistent with our recent work, male, but not female, mice displayed behavioral and cognitive deficits with corresponding perturbations of neuroimmune factors in the PFC after 14 days of CUS. Fluorescence-activated cell sorting and gene expression analyses revealed that CUS increased expression of select markers of phagocytosis in male PFC microglia. Confocal imaging in Thy1-GFP(M) mice showed that CUS reduced dendritic spine density, decreased GFAP immunolabeling, and increased microglia-mediated neuronal remodeling only in male mice. After 28 days of CUS, both male and female mice displayed behavioral and cognitive impairments. Interestingly, there were limited stress effects on neuroimmune factors and measures of microglial phagocytosis in the PFC of both sexes. Despite limited changes in neuroimmune function, reduced GFAP immunolabeling and dendritic spine deficits persisted in male mice. Further, GFAP immunolabeling and dendritic spine density remained unaltered in the PFC of females. These findings indicate that chronic stress causes sex-specific and temporally dynamic changes in microglial function which are associated with different neurobiological and behavioral adaptations. In all, these results suggest that microglia-mediated neuronal remodeling, astrocyte dystrophy, and synapse loss contribute to stress-induced PFC dysfunction and associated behavioral consequences in male mice.
Collapse
Affiliation(s)
- Samuel C. Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin L. Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S. Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Corresponding author. Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, 2120 East Galbraith Road, Cincinnati, OH, 45237, USA.
| |
Collapse
|
276
|
Guttikonda SR, Sikkema L, Tchieu J, Saurat N, Walsh R, Harschnitz O, Ciceri G, Sneeboer M, Mazutis L, Setty M, Zumbo P, Betel D, de Witte LD, Pe’er D, Studer L. Fully defined human pluripotent stem cell-derived microglia and tri-culture system model C3 production in Alzheimer's disease. Nat Neurosci 2021; 24:343-354. [PMID: 33558694 PMCID: PMC8382543 DOI: 10.1038/s41593-020-00796-z] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 12/24/2020] [Indexed: 01/30/2023]
Abstract
Aberrant inflammation in the CNS has been implicated as a major player in the pathogenesis of human neurodegenerative disease. We developed a new approach to derive microglia from human pluripotent stem cells (hPSCs) and built a defined hPSC-derived tri-culture system containing pure populations of hPSC-derived microglia, astrocytes, and neurons to dissect cellular cross-talk along the neuroinflammatory axis in vitro. We used the tri-culture system to model neuroinflammation in Alzheimer's disease with hPSCs harboring the APPSWE+/+ mutation and their isogenic control. We found that complement C3, a protein that is increased under inflammatory conditions and implicated in synaptic loss, is potentiated in tri-culture and further enhanced in APPSWE+/+ tri-cultures due to microglia initiating reciprocal signaling with astrocytes to produce excess C3. Our study defines the major cellular players contributing to increased C3 in Alzheimer's disease and presents a broadly applicable platform to study neuroinflammation in human disease.
Collapse
Affiliation(s)
- Sudha R. Guttikonda
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065 USA
| | - Lisa Sikkema
- Computational and Systems Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Metastasis & Tumor Ecosystems Center, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA
| | - Jason Tchieu
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA
| | - Nathalie Saurat
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA
| | - Ryan Walsh
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA
| | - Oliver Harschnitz
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA
| | - Gabriele Ciceri
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA
| | - Marjolein Sneeboer
- Institute for Computational Biomedicine, Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, New York, NY 10065, USA
| | - Linas Mazutis
- Computational and Systems Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Metastasis & Tumor Ecosystems Center, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA
| | - Manu Setty
- Computational and Systems Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Metastasis & Tumor Ecosystems Center, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA
| | - Paul Zumbo
- Applied Bioinformatics Core & Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, New York, NY 10065, USA
| | - Doron Betel
- Institute for Computational Biomedicine, Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, New York, NY 10065, USA
| | - Lot D. de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY Mount Sinai Medical Center,Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Dana Pe’er
- Computational and Systems Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Metastasis & Tumor Ecosystems Center, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY 10065 USA,Correspondence to: Dr. Lorenz Studer, The Center for Stem Cell Biology, Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Box 256, New York, NY 10065, Phone: 212-639-6126,
| |
Collapse
|
277
|
Shinjyo N, Nakayama H, Li L, Ishimaru K, Hikosaka K, Suzuki N, Yoshida H, Norose K. Hypericum perforatum extract and hyperforin inhibit the growth of neurotropic parasite Toxoplasma gondii and infection-induced inflammatory responses of glial cells in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113525. [PMID: 33129946 DOI: 10.1016/j.jep.2020.113525] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/19/2020] [Accepted: 10/26/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hypericum perforatum L. has been widely used as a natural antidepressant. However, it is unknown whether it is effective in treating infection-induced neuropsychiatric disorders. AIM OF THE STUDY In order to evaluate the effectiveness of H. perforatum against infection with neurotropic parasite Toxoplasma gondii, which has been linked to neuropsychiatric disorders, this study investigated the anti-Toxoplasma activity using in vitro models. MATERIALS AND METHODS Dried alcoholic extracts were prepared from three Hypericum species: H. perforatum, H. erectum, and H. ascyron. H. perforatum extract was further separated by solvent-partitioning. Hyperforin and hypericin levels in the extracts and fractions were analyzed by high resolution LC-MS. Anti-Toxoplasma activities were tested in vitro, using cell lines (Vero and Raw264), murine primary mixed glia, and primary neuron-glia. Toxoplasma proliferation and stage conversion were analyzed by qPCR. Infection-induced damages to the host cells were analyzed by Sulforhodamine B cytotoxicity assay (Vero) and immunofluorescent microscopy (neurons). Infection-induced inflammatory responses in glial cells were analysed by qPCR and immunofluorescent microscopy. RESULTS Hyperforin was identified only in H. perforatum among the three tested species, whereas hypericin was present in H. perforatum and H. erectum. H. perforatum extract and hyperforin-enriched fraction, as well as hyperforin, exhibited significant anti-Toxoplasma property as well as inhibitory activity against infection-induced inflammatory responses in glial cells. In addition, H. perforatum-derived hyperforin-enriched fraction restored neuro-supportive environment in mixed neuron-glia culture. CONCLUSIONS H. perforatum and its major constituent hyperforin are promising anti-Toxoplasma agents that could potentially protect neurons and glial cells against infection-induced damages. Further study is warranted to establish in vivo efficacy.
Collapse
Affiliation(s)
- Noriko Shinjyo
- Department of Infection and Host Defense, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan; School of Tropical Medicine and Global Health, Nagasaki University, 1-12-4, Sakamoto, Nagasaki, 852-8523, Japan.
| | - Hideyuki Nakayama
- Saga Prefectural Institute of Public Health and Pharmaceutical Research, 1-20 Hacchounawate, Saga, 849-0925, Japan
| | - Li Li
- Department of Infection and Host Defense, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Kanji Ishimaru
- Department of Biological Resource Sciences, Faculty of Agriculture, Saga University, 1 Honjo, Saga, 840-8502, Japan
| | - Kenji Hikosaka
- Department of Infection and Host Defense, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Noriyuki Suzuki
- Department of Toxicology and Environmental Health, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Kazumi Norose
- Department of Infection and Host Defense, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| |
Collapse
|
278
|
Cardiac glycosides target barrier inflammation of the vasculature, meninges and choroid plexus. Commun Biol 2021; 4:260. [PMID: 33637884 PMCID: PMC7910294 DOI: 10.1038/s42003-021-01787-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Neuroinflammation is a key component of virtually all neurodegenerative diseases, preceding neuronal loss and associating directly with cognitive impairment. Neuroinflammatory signals can originate and be amplified at barrier tissues such as brain vasculature, surrounding meninges and the choroid plexus. We designed a high content screening system to target inflammation in human brain-derived cells of the blood-brain barrier (pericytes and endothelial cells) to identify inflammatory modifiers. Screening an FDA-approved drug library we identify digoxin and lanatoside C, members of the cardiac glycoside family, as inflammatory-modulating drugs that work in blood-brain barrier cells. An ex vivo assay of leptomeningeal and choroid plexus explants confirm that these drugs maintain their function in 3D cultures of brain border tissues. These results suggest that cardiac glycosides may be useful in targeting inflammation at border regions of the brain and offer new options for drug discovery approaches for neuroinflammatory driven degeneration.
Collapse
|
279
|
Monitoring and Modulating Inflammation-Associated Alterations in Synaptic Plasticity: Role of Brain Stimulation and the Blood-Brain Interface. Biomolecules 2021; 11:biom11030359. [PMID: 33652912 PMCID: PMC7996828 DOI: 10.3390/biom11030359] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 12/20/2022] Open
Abstract
Inflammation of the central nervous system can be triggered by endogenous and exogenous stimuli such as local or systemic infection, trauma, and stroke. In addition to neurodegeneration and cell death, alterations in physiological brain functions are often associated with neuroinflammation. Robust experimental evidence has demonstrated that inflammatory cytokines affect the ability of neurons to express plasticity. It has been well-established that inflammation-associated alterations in synaptic plasticity contribute to the development of neuropsychiatric symptoms. Nevertheless, diagnostic approaches and interventional strategies to restore inflammatory deficits in synaptic plasticity are limited. Here, we review recent findings on inflammation-associated alterations in synaptic plasticity and the potential role of the blood–brain interface, i.e., the blood–brain barrier, in modulating synaptic plasticity. Based on recent findings indicating that brain stimulation promotes plasticity and modulates vascular function, we argue that clinically employed non-invasive brain stimulation techniques, such as transcranial magnetic stimulation, could be used for monitoring and modulating inflammation-induced alterations in synaptic plasticity.
Collapse
|
280
|
Loss of NPC1 enhances phagocytic uptake and impairs lipid trafficking in microglia. Nat Commun 2021; 12:1158. [PMID: 33627648 PMCID: PMC7904859 DOI: 10.1038/s41467-021-21428-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/27/2021] [Indexed: 02/08/2023] Open
Abstract
Niemann-Pick type C disease is a rare neurodegenerative disorder mainly caused by mutations in NPC1, resulting in abnormal late endosomal/lysosomal lipid storage. Although microgliosis is a prominent pathological feature, direct consequences of NPC1 loss on microglial function remain not fully characterized. We discovered pathological proteomic signatures and phenotypes in NPC1-deficient murine models and demonstrate a cell autonomous function of NPC1 in microglia. Loss of NPC1 triggers enhanced phagocytic uptake and impaired myelin turnover in microglia that precede neuronal death. Npc1−/− microglia feature a striking accumulation of multivesicular bodies and impaired trafficking of lipids to lysosomes while lysosomal degradation function remains preserved. Molecular and functional defects were also detected in blood-derived macrophages of NPC patients that provide a potential tool for monitoring disease. Our study underscores an essential cell autonomous role for NPC1 in immune cells and implies microglial therapeutic potential. Niemann-Pick type C disease is a rare childhood neurodegenerative disorder predominantly caused by mutations in NPC1, resulting in abnormal late endosomal and lysosomal defects. Here the authors show that NPC1 disruption largely impairs microglial function.
Collapse
|
281
|
Morini R, Bizzotto M, Perrucci F, Filipello F, Matteoli M. Strategies and Tools for Studying Microglial-Mediated Synapse Elimination and Refinement. Front Immunol 2021; 12:640937. [PMID: 33708226 PMCID: PMC7940197 DOI: 10.3389/fimmu.2021.640937] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/01/2021] [Indexed: 01/14/2023] Open
Abstract
The role of microglia in controlling synapse homeostasis is becoming increasingly recognized by the scientific community. In particular, the microglia-mediated elimination of supernumerary synapses during development lays the basis for the correct formation of neuronal circuits in adulthood, while the possible reactivation of this process in pathological conditions, such as schizophrenia or Alzheimer's Disease, provides a promising target for future therapeutic strategies. The methodological approaches to investigate microglial synaptic engulfment include different in vitro and in vivo settings. Basic in vitro assays, employing isolated microglia and microbeads, apoptotic membranes, liposomes or synaptosomes allow the quantification of the microglia phagocytic abilities, while co-cultures of microglia and neurons, deriving from either WT or genetically modified mice models, provide a relatively manageable setting to investigate the involvement of specific molecular pathways. Further detailed analysis in mice brain is then mandatory to validate the in vitro assays as representative for the in vivo situation. The present review aims to dissect the main technical approaches to investigate microglia-mediated phagocytosis of neuronal and synaptic substrates in critical developmental time windows.
Collapse
Affiliation(s)
- Raffaella Morini
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Matteo Bizzotto
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Fabio Perrucci
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Fabia Filipello
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Consiglio Nazionale Delle Ricerche (CNR), Institute of Neuroscience - URT Humanitas, Rozzano, Italy
| |
Collapse
|
282
|
Askenase MH, Goods BA, Beatty HE, Steinschneider AF, Velazquez SE, Osherov A, Landreneau MJ, Carroll SL, Tran TB, Avram VS, Drake RS, Gatter GJ, Massey JA, Karuppagounder SS, Ratan RR, Matouk CC, Sheth KN, Ziai WC, Parry-Jones AR, Awad IA, Zuccarello M, Thompson RE, Dawson J, Hanley DF, Love JC, Shalek AK, Sansing LH. Longitudinal transcriptomics define the stages of myeloid activation in the living human brain after intracerebral hemorrhage. Sci Immunol 2021; 6:6/56/eabd6279. [PMID: 33891558 DOI: 10.1126/sciimmunol.abd6279] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/21/2021] [Indexed: 12/20/2022]
Abstract
Opportunities to interrogate the immune responses in the injured tissue of living patients suffering from acute sterile injuries such as stroke and heart attack are limited. We leveraged a clinical trial of minimally invasive neurosurgery for patients with intracerebral hemorrhage (ICH), a severely disabling subtype of stroke, to investigate the dynamics of inflammation at the site of brain injury over time. Longitudinal transcriptional profiling of CD14+ monocytes/macrophages and neutrophils from hematomas of patients with ICH revealed that the myeloid response to ICH within the hematoma is distinct from that in the blood and occurs in stages conserved across the patient cohort. Initially, hematoma myeloid cells expressed a robust anabolic proinflammatory profile characterized by activation of hypoxia-inducible factors (HIFs) and expression of genes encoding immune factors and glycolysis. Subsequently, inflammatory gene expression decreased over time, whereas anti-inflammatory circuits were maintained and phagocytic and antioxidative pathways up-regulated. During this transition to immune resolution, glycolysis gene expression and levels of the potent proresolution lipid mediator prostaglandin E2 remained elevated in the hematoma, and unexpectedly, these elevations correlated with positive patient outcomes. Ex vivo activation of human macrophages by ICH-associated stimuli highlighted an important role for HIFs in production of both inflammatory and anti-inflammatory factors, including PGE2, which, in turn, augmented VEGF production. Our findings define the time course of myeloid activation in the human brain after ICH, revealing a conserved progression of immune responses from proinflammatory to proresolution states in humans after brain injury and identifying transcriptional programs associated with neurological recovery.
Collapse
Affiliation(s)
- Michael H Askenase
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Brittany A Goods
- Institute for Medical Engineering & Science (IMES) and Department of Chemistry, MIT, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Hannah E Beatty
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Arthur F Steinschneider
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Sofia E Velazquez
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Artem Osherov
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Margaret J Landreneau
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Shaina L Carroll
- Institute for Medical Engineering & Science (IMES) and Department of Chemistry, MIT, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Tho B Tran
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Victor S Avram
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Riley S Drake
- Institute for Medical Engineering & Science (IMES) and Department of Chemistry, MIT, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - G James Gatter
- Institute for Medical Engineering & Science (IMES) and Department of Chemistry, MIT, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Jordan A Massey
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Saravanan S Karuppagounder
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Rajiv R Ratan
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Charles C Matouk
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Kevin N Sheth
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Wendy C Ziai
- Division of Brain Injury Outcomes, Johns Hopkins University, Baltimore, MD, USA.,Departments of Neurology, Neurosurgery, and Anesthesiology/Critical Care Medicine, Johns Hopkins, Baltimore, MD, USA
| | - Adrian R Parry-Jones
- Division of Cardiovascular Sciences, School of Medicine, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.,Manchester Centre for Clinical Neurosciences, Salford Royal National Health Service Foundation Trust, Manchester Academic Health Science Centre, Salford, UK
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Mario Zuccarello
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Richard E Thompson
- Division of Brain Injury Outcomes, Johns Hopkins University, Baltimore, MD, USA.,Department of Biostatistics, School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Jesse Dawson
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Daniel F Hanley
- Division of Brain Injury Outcomes, Johns Hopkins University, Baltimore, MD, USA
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Alex K Shalek
- Institute for Medical Engineering & Science (IMES) and Department of Chemistry, MIT, Cambridge, MA, USA. .,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Lauren H Sansing
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA. .,Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.,Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT, USA
| | | | | | | |
Collapse
|
283
|
Abstract
Chronic neuroinflammation is observed in HIV+ individuals on suppressive combination antiretroviral therapy (cART) and is thought to cause HIV-associated neurocognitive disorders. We have recently reported that expression of HIV intron-containing RNA (icRNA) in productively infected monocyte-derived macrophages induces pro-inflammatory responses. Microglia, yolk sac-derived brain-resident tissue macrophages, are the primary HIV-1 infected cell type in the central nervous system (CNS). In this study, we tested the hypothesis that persistent expression of HIV icRNA in primary human microglia induces innate immune activation. We established multiple orthogonal primary human microglia-like cell cultures including peripheral blood monocyte-derived microglia (MDMG) and induced pluripotent stem cell (iPSC)-derived microglia. Unlike MDMG, human iPSC-derived microglia (hiMG), which phenotypically mimic primary CNS microglia, were robustly infected with replication competent HIV-1, and establishment of productive HIV-1 infection and de novo viral gene expression led to pro-inflammatory cytokine production. Blocking of HIV-1 icRNA expression, but not multiply spliced viral RNA, either via infection with virus expressing a Rev-mutant deficient for HIV icRNA nuclear export or infection in the presence of small molecule inhibitor of CRM1-mediated viral icRNA nuclear export pathway, attenuated induction of innate immune responses. These studies suggest that Rev-CRM1-dependent nuclear export and cytosolic sensing of HIV-1 icRNA induces pro-inflammatory responses in productively infected microglia. Novel strategies targeting HIV icRNA expression specifically are needed to suppress HIV-induced neuroinflammation.
Collapse
|
284
|
Delaney C, Farrell M, Doherty CP, Brennan K, O’Keeffe E, Greene C, Byrne K, Kelly E, Birmingham N, Hickey P, Cronin S, Savvides SN, Doyle SL, Campbell M. Attenuated CSF-1R signalling drives cerebrovascular pathology. EMBO Mol Med 2021; 13:e12889. [PMID: 33350588 PMCID: PMC7863388 DOI: 10.15252/emmm.202012889] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
Cerebrovascular pathologies occur in up to 80% of cases of Alzheimer's disease; however, the underlying mechanisms that lead to perivascular pathology and accompanying blood-brain barrier (BBB) disruption are still not fully understood. We have identified previously unreported mutations in colony stimulating factor-1 receptor (CSF-1R) in an ultra-rare autosomal dominant condition termed adult-onset leucoencephalopathy with axonal spheroids and pigmented glia (ALSP). Cerebrovascular pathologies such as cerebral amyloid angiopathy (CAA) and perivascular p-Tau were some of the primary neuropathological features of this condition. We have identified two families with different dominant acting alleles with variants located in the kinase region of the CSF-1R gene, which confer a lack of kinase activity and signalling. The protein product of this gene acts as the receptor for 2 cognate ligands, namely colony stimulating factor-1 (CSF-1) and interleukin-34 (IL-34). Here, we show that depletion in CSF-1R signalling induces BBB disruption and decreases the phagocytic capacity of peripheral macrophages but not microglia. CSF-1R signalling appears to be critical for macrophage and microglial activation, and macrophage localisation to amyloid appears reduced following the induction of Csf-1r heterozygosity in macrophages. Finally, we show that endothelial/microglial crosstalk and concomitant attenuation of CSF-1R signalling causes re-modelling of BBB-associated tight junctions and suggest that regulating BBB integrity and systemic macrophage recruitment to the brain may be therapeutically relevant in ALSP and other Alzheimer's-like dementias.
Collapse
Affiliation(s)
- Conor Delaney
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Michael Farrell
- Department of NeuropathologyBeaumont HospitalDublin 9Ireland
| | - Colin P Doherty
- Department of NeurologyHealth Care CentreSt James's HospitalDublin 8Ireland
- Academic Unit of NeurologyBiomedical Sciences InstituteTrinity College DublinDublin 2Ireland
- FutureNeuro SFI Research CentreRoyal College of Surgeons in IrelandDublinIreland
| | - Kiva Brennan
- Trinity College Institute of NeuroscienceTrinity College Dublin 2Dublin 2Ireland
| | - Eoin O’Keeffe
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Chris Greene
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Kieva Byrne
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Eoin Kelly
- Department of NeurologyHealth Care CentreSt James's HospitalDublin 8Ireland
| | | | | | - Simon Cronin
- Department of MedicineUniversity College CorkCorkIreland
| | - Savvas N Savvides
- Unit for Structural BiologyDepartment of Biochemistry and MicrobiologyGhent UniversityGhentBelgium
- VIB‐UGent Center for Inflammation ResearchGhentBelgium
| | - Sarah L Doyle
- Trinity College Institute of NeuroscienceTrinity College Dublin 2Dublin 2Ireland
| | - Matthew Campbell
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
- FutureNeuro SFI Research CentreRoyal College of Surgeons in IrelandDublinIreland
| |
Collapse
|
285
|
Astrocyte-immune cell interactions in physiology and pathology. Immunity 2021; 54:211-224. [DOI: 10.1016/j.immuni.2021.01.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/29/2020] [Accepted: 01/15/2021] [Indexed: 12/23/2022]
|
286
|
Does tissue imprinting restrict macrophage plasticity? Nat Immunol 2021; 22:118-127. [PMID: 33462453 DOI: 10.1038/s41590-020-00849-2] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 11/20/2020] [Indexed: 01/29/2023]
Abstract
Macrophages have long been considered as particularly plastic cells. However, recent work combining fate mapping, single-cell transcriptomics and epigenetics has undermined the macrophage plasticity dogma. Here, we discuss recent studies that have carefully dissected the response of individual macrophage subsets to pulmonary insults and call for an adjustment of the macrophage plasticity concept. We hypothesize that prolonged tissue residency shuts down much of the plasticity of macrophages and propose that the restricted plasticity of resident macrophages has been favored by evolution to safeguard tissue homeostasis. Recruited monocytes are more plastic and their differentiation into resident macrophages during inflammation can result in a dual imprinting from both the ongoing inflammation and the macrophage niche. This results in inflammation-imprinted resident macrophages, and we speculate that rewired niche circuits could maintain this inflammatory state. We believe that this revisited plasticity model offers opportunities to reset the macrophage pool after a severe inflammatory episode.
Collapse
|
287
|
Miller-Rhodes P, Gelbard HA. The Cell Culture Environment Regulates the Transcription Factor MafB in BV-2 Microglia. MATTERS 2021; 2021:https://sciencematters.io/articles/202010000001. [PMID: 33969051 PMCID: PMC8101704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Microglia experience dramatic molecular and functional changes when transferred from the central nervous system (CNS) to a cell culture environment. Investigators largely attribute these findings to the loss of CNS-specific microenvironmental cues that dictate the gene-regulatory networks specified by master regulator transcription factors such as V-maf musculoaponeurotic fibrosarcoma oncogene homolog B (MafB). MafB regulates macrophage differentiation and activation by activating or repressing target genes critical to these processes. Here, we show that basal MafB levels in the BV-2 microglial cell line depend on the availability of lipids in the cell culture environment. Depletion of lipids, either by serum deprivation or the use of lipid-depleted serum, reduced MafB protein levels in BV-2 cells. Using live imaging, we also observed the engulfment of apoptotic BV-2 cell debris by neighboring BV-2 cells, highlighting an additional potential source of lipids in the cell culture environment. This observation was supported by experiments showing reduced MafB protein levels in BV-2 cells cultured with various phagocytosis inhibitors (cytochalasin D, annexin V) and reduced BV-2 cell phagocytic activity with serum deprivation. In aggregate, our data suggest that serum exposure regulates the transcription factor MafB in BV-2 cells through direct and indirect mechanisms.
Collapse
Affiliation(s)
- Patrick Miller-Rhodes
- Center for Neurotherapeutics Discovery, Department of Neuroscience, Department of Immunology and Microbiology, Department of Neurology, Department of Pediatrics, University of Rochester Medical Center
| | - Harris A Gelbard
- Center for Neurotherapeutics Discovery, Department of Neuroscience, Department of Immunology and Microbiology, Department of Neurology, Department of Pediatrics, University of Rochester Medical Center
| |
Collapse
|
288
|
Bart VMT, Pickering RJ, Taylor PR, Ipseiz N. Macrophage reprogramming for therapy. Immunology 2021; 163:128-144. [PMID: 33368269 PMCID: PMC8114216 DOI: 10.1111/imm.13300] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/02/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Dysfunction of the immune system underlies a plethora of human diseases, requiring the development of immunomodulatory therapeutic intervention. To date, most strategies employed have been focusing on the modification of T lymphocytes, and although remarkable improvement has been obtained, results often fall short of the intended outcome. Recent cutting-edge technologies have highlighted macrophages as potential targets for disease control. Macrophages play central roles in development, homeostasis and host defence, and their dysfunction and dysregulation have been implicated in the onset and pathogenesis of multiple disorders including cancer, neurodegeneration, autoimmunity and metabolic diseases. Recent advancements have led to a greater understanding of macrophage origin, diversity and function, in both health and disease. Over the last few years, a variety of strategies targeting macrophages have been developed and these open new therapeutic opportunities. Here, we review the progress in macrophage reprogramming in various disorders and discuss the potential implications and challenges for macrophage-targeted approaches in human disease.
Collapse
Affiliation(s)
| | - Robert J Pickering
- Immunology Network, Adaptive Immunity Research Unit, GlaxoSmithKline, Stevenage, UK.,Department of Medicine, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Philip R Taylor
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK.,UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, UK
| | - Natacha Ipseiz
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
289
|
Fattorelli N, Martinez-Muriana A, Wolfs L, Geric I, De Strooper B, Mancuso R. Stem-cell-derived human microglia transplanted into mouse brain to study human disease. Nat Protoc 2021; 16:1013-1033. [PMID: 33424025 DOI: 10.1038/s41596-020-00447-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/13/2020] [Indexed: 11/09/2022]
Abstract
Microglia are critically involved in complex neurological disorders with a strong genetic component, such as Alzheimer's disease, Parkinson's disease and frontotemporal dementia. Although mouse microglia can recapitulate aspects of human microglia physiology, they do not fully capture the human genetic aspects of disease and do not reproduce all human cell states. Primary cultures of human microglia or microglia derived from human induced pluripotent stem cells (PSCs) are difficult to maintain in brain-relevant cell states in vitro. Here we describe MIGRATE (microglia in vitro generation refined for advanced transplantation experiments, which provides a combined in vitro differentiation and in vivo xenotransplantation protocol to study human microglia in the context of the mouse brain. This article details an accurate, step-by-step workflow that includes in vitro microglia differentiation from human PSCs, transplantation into the mouse brain and quantitative analysis of engraftment. Compared to current differentiation and xenotransplantation protocols, we present an optimized, faster and more efficient approach that yields up to 80% chimerism. To quantitatively assess engraftment efficiency by flow cytometry, access to specialized flow cytometry is required. Alternatively, the percentage of chimerism can be estimated by standard immunohistochemical analysis. The MIGRATE protocol takes ~40 d to complete, from culturing PSCs to engraftment efficiency assessment.
Collapse
Affiliation(s)
- Nicola Fattorelli
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium.,Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Anna Martinez-Muriana
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium.,Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Leen Wolfs
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium.,Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Ivana Geric
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium.,Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium. .,Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium. .,UK Dementia Research Institute at UCL, University College London, London, UK.
| | - Renzo Mancuso
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium. .,Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium. .,Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium. .,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
290
|
Fluid Biomarkers of Frontotemporal Lobar Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:123-139. [PMID: 33433873 DOI: 10.1007/978-3-030-51140-1_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A timely diagnosis of frontotemporal degeneration (FTD) is frequently challenging due to the heterogeneous symptomatology and poor phenotype-pathological correlation. Fluid biomarkers that reflect FTD pathophysiology could be instrumental in both clinical practice and pharmaceutical trials. In recent years, significant progress has been made in developing biomarkers of neurodegenerative diseases: amyloid-β and tau in cerebrospinal fluid (CSF) can be used to exclude Alzheimer's disease, while neurofilament light chain (NfL) is emerging as a promising, albeit nonspecific, marker of neurodegeneration in both CSF and blood. Gene-specific biomarkers such as PGRN in GRN mutation carriers and dipeptide repeat proteins in C9orf72 mutation carriers are potential target engagement markers in genetic FTD trials. Novel techniques capable of measuring very low concentrations of brain-derived proteins in peripheral fluids are facilitating studies of blood biomarkers as a minimally invasive alternative to CSF. A major remaining challenge is the identification of a biomarker that can be used to predict the neuropathological substrate in sporadic FTD patients.
Collapse
|
291
|
Abstract
A major feature of neurodegeneration is disruption of central nervous system homeostasis, during which microglia play diverse roles. In the central nervous system, microglia serve as the first line of immune defense and function in synapse pruning, injury repair, homeostasis maintenance, and regulation of brain development through scavenging and phagocytosis. Under pathological conditions or various stimulations, microglia proliferate, aggregate, and undergo a variety of changes in cell morphology, immunophenotype, and function. This review presents the features of microglia, especially their diversity and ability to change dynamically, and reinterprets their role as sensors for multiple stimulations and as effectors for brain aging and neurodegeneration. This review also summarizes some therapeutic approaches for neurodegenerative diseases that target microglia.
Collapse
Affiliation(s)
- Yu Xu
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Municipal Key Clinical Specialty; Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Zhu Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Yong Yang
- Department of Anesthesiology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Municipal Key Clinical Specialty, Shanghai, China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering; National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai; Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
292
|
Bassett B, Subramaniyam S, Fan Y, Varney S, Pan H, Carneiro AMD, Chung CY. Minocycline alleviates depression-like symptoms by rescuing decrease in neurogenesis in dorsal hippocampus via blocking microglia activation/phagocytosis. Brain Behav Immun 2021; 91:519-530. [PMID: 33176182 DOI: 10.1016/j.bbi.2020.11.009] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/12/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Clinical studies examining the potential of anti-inflammatory agents, specifically of minocycline, as a treatment for depression has shown promising results. However, mechanistic insights into the neuroprotective and anti-inflammatory actions of minocycline need to be provided. We evaluated the effect of minocycline on chronic mild stress (CMS) induced depressive-like behavior, and behavioral assays revealed minocycline ameliorate depressive behaviors. Multiple studies suggest a role of microglia in depression, revealing that microglia activation correlates with a decrease in neurogenesis and increased depressive-like behavior. The effect of minocycline on microglia activation in different areas of the dorsal or ventral hippocampus in stressed mice was examined by immunohistochemistry. We observed the increase in the number of activated microglia expressing CD68 after exposure to three weeks of chronic stress, whereas no changes in total microglia number were observed. These changes were observed throughout the DG, CA1 and CA2 regions in dorsal hippocampus but restricted to the DG of the ventral hippocampus. In vitro experiments including western blotting and phagocytosis assay were used to investigate the effect of minocycline on microglia activation. Activation of primary microglia by LPS in vitro causes and ERK1/2 activation, enhancement of iNOS expression and phagocytic activity, and alterations in cellular morphology that are reversed by minocycline exposure, suggesting that minocycline directly acts on microglia to reduce phagocytic potential. Our results suggest the most probable mechanism by which minocycline reverses the pathogenic phagocytic potential of neurotoxic M1 microglia, and reduces the negative phenotypes associated with reduced neurogenesis caused by exposure to chronic stress.
Collapse
Affiliation(s)
- Ben Bassett
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Selvaraj Subramaniyam
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yang Fan
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Seth Varney
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Hope Pan
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Ana M D Carneiro
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Chang Y Chung
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China; Division of Natural Science, Duke Kunshan University, Kunshan 215316, China.
| |
Collapse
|
293
|
Cunha MI, Su M, Cantuti-Castelvetri L, Müller SA, Schifferer M, Djannatian M, Alexopoulos I, van der Meer F, Winkler A, van Ham TJ, Schmid B, Lichtenthaler SF, Stadelmann C, Simons M. Pro-inflammatory activation following demyelination is required for myelin clearance and oligodendrogenesis. J Exp Med 2020; 217:133824. [PMID: 32078678 PMCID: PMC7201919 DOI: 10.1084/jem.20191390] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 11/22/2019] [Accepted: 01/17/2020] [Indexed: 12/19/2022] Open
Abstract
Remyelination requires innate immune system function, but how exactly microglia and macrophages clear myelin debris after injury and tailor a specific regenerative response is unclear. Here, we asked whether pro-inflammatory microglial/macrophage activation is required for this process. We established a novel toxin-based spinal cord model of de- and remyelination in zebrafish and showed that pro-inflammatory NF-κB–dependent activation in phagocytes occurs rapidly after myelin injury. We found that the pro-inflammatory response depends on myeloid differentiation primary response 88 (MyD88). MyD88-deficient mice and zebrafish were not only impaired in the degradation of myelin debris, but also in initiating the generation of new oligodendrocytes for myelin repair. We identified reduced generation of TNF-α in lesions of MyD88-deficient animals, a pro-inflammatory molecule that was able to induce the generation of new premyelinating oligodendrocytes. Our study shows that pro-inflammatory phagocytic signaling is required for myelin debris degradation, for inflammation resolution, and for initiating the generation of new oligodendrocytes.
Collapse
Affiliation(s)
- Maria Inês Cunha
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany.,Graduate Program in Areas of Basic and Applied Biology, Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - Minhui Su
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany
| | - Ludovico Cantuti-Castelvetri
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany
| | | | - Martina Schifferer
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany
| | - Minou Djannatian
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany
| | - Ioannis Alexopoulos
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany
| | - Franziska van der Meer
- Department of Neuropathology, University of Göttingen Medical Center, Göttingen, Germany
| | - Anne Winkler
- Department of Neuropathology, University of Göttingen Medical Center, Göttingen, Germany
| | - Tjakko J van Ham
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Bettina Schmid
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases, Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University of Göttingen Medical Center, Göttingen, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.,Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
294
|
Lenz M, Eichler A, Kruse P, Strehl A, Rodriguez-Rozada S, Goren I, Yogev N, Frank S, Waisman A, Deller T, Jung S, Maggio N, Vlachos A. Interleukin 10 Restores Lipopolysaccharide-Induced Alterations in Synaptic Plasticity Probed by Repetitive Magnetic Stimulation. Front Immunol 2020; 11:614509. [PMID: 33391287 PMCID: PMC7772211 DOI: 10.3389/fimmu.2020.614509] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/16/2020] [Indexed: 12/26/2022] Open
Abstract
Systemic inflammation is associated with alterations in complex brain functions such as learning and memory. However, diagnostic approaches to functionally assess and quantify inflammation-associated alterations in synaptic plasticity are not well-established. In previous work, we demonstrated that bacterial lipopolysaccharide (LPS)-induced systemic inflammation alters the ability of hippocampal neurons to express synaptic plasticity, i.e., the long-term potentiation (LTP) of excitatory neurotransmission. Here, we tested whether synaptic plasticity induced by repetitive magnetic stimulation (rMS), a non-invasive brain stimulation technique used in clinical practice, is affected by LPS-induced inflammation. Specifically, we explored brain tissue cultures to learn more about the direct effects of LPS on neural tissue, and we tested for the plasticity-restoring effects of the anti-inflammatory cytokine interleukin 10 (IL10). As shown previously, 10 Hz repetitive magnetic stimulation (rMS) of organotypic entorhino-hippocampal tissue cultures induced a robust increase in excitatory neurotransmission onto CA1 pyramidal neurons. Furthermore, LPS-treated tissue cultures did not express rMS-induced synaptic plasticity. Live-cell microscopy in tissue cultures prepared from a novel transgenic reporter mouse line [C57BL/6-Tg(TNFa-eGFP)] confirms that ex vivo LPS administration triggers microglial tumor necrosis factor alpha (TNFα) expression, which is ameliorated in the presence of IL10. Consistent with this observation, IL10 hampers the LPS-induced increase in TNFα, IL6, IL1β, and IFNγ and restores the ability of neurons to express rMS-induced synaptic plasticity in the presence of LPS. These findings establish organotypic tissue cultures as a suitable model for studying inflammation-induced alterations in synaptic plasticity, thus providing a biological basis for the diagnostic use of transcranial magnetic stimulation in the context of brain inflammation.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Strehl
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt, Germany
| | - Silvia Rodriguez-Rozada
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt, Germany
| | - Itamar Goren
- Pharmazentrum Frankfurt/ZAFES, Goethe-University Frankfurt, Frankfurt, Germany
| | - Nir Yogev
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Dermatology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Stefan Frank
- Pharmazentrum Frankfurt/ZAFES, Goethe-University Frankfurt, Frankfurt, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt, Germany
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Nicola Maggio
- Department of Neurology and Sagol Center for Neurosciences, Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Tel HaShomer, Israel
- Sackler Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
295
|
Bruzelius A, Hidalgo I, Boza-Serrano A, Hjelmér AG, Tison A, Deierborg T, Bengzon J, Ramos-Moreno T. The human bone marrow harbors a CD45 - CD11B + cell progenitor permitting rapid microglia-like cell derivative approaches. Stem Cells Transl Med 2020; 10:582-597. [PMID: 33295698 PMCID: PMC7980218 DOI: 10.1002/sctm.20-0127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 09/23/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022] Open
Abstract
Microglia, the immune sentinel of the central nervous system (CNS), are generated from yolk sac erythromyeloid progenitors that populate the developing CNS. Interestingly, a specific type of bone marrow-derived monocyte is able to express a yolk sac microglial signature and populate CNS in disease. Here we have examined human bone marrow (hBM) in an attempt to identify novel cell sources for generating microglia-like cells to use in cell-based therapies and in vitro modeling. We demonstrate that hBM stroma harbors a progenitor cell that we name stromal microglial progenitor (STR-MP). STR-MP single-cell gene analysis revealed the expression of the consensus genetic microglial signature and microglial-specific genes present in development and CNS pathologies. STR-MPs can be expanded and generate microglia-like cells in vitro, which we name stromal microglia (STR-M). STR-M cells show phagocytic ability, classically activate, and survive and phagocyte in human brain tissue. Thus, our results reveal that hBM harbors a source of microglia-like precursors that can be used in patient-centered fast derivative approaches.
Collapse
Affiliation(s)
- Andreas Bruzelius
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden.,Department of Experimental Medical Science and Lund Stem Cell Center BMC, Lund University, Lund, Sweden
| | - Isabel Hidalgo
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden.,Institution for Laboratory Medicine, Division of Molecular Hematology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Antonio Boza-Serrano
- Departamento de Bioquimica y Biologia Molecular, Facultad de Farmacia e Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain.,Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Bio Medical Center (BMC)., Lund University, Lund, Sweden
| | - Anna-Giorgia Hjelmér
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden
| | - Amelie Tison
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Bio Medical Center (BMC)., Lund University, Lund, Sweden
| | - Johan Bengzon
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden
| | - Tania Ramos-Moreno
- Division of Neurosurgery, Department of Clinical Sciences Lund, Skåne University Hospital, Lund Stem Cell Center, Lund, Sweden.,Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Bio Medical Center (BMC)., Lund University, Lund, Sweden
| |
Collapse
|
296
|
Hedegaard A, Stodolak S, James WS, Cowley SA. Honing the Double-Edged Sword: Improving Human iPSC-Microglia Models. Front Immunol 2020; 11:614972. [PMID: 33363548 PMCID: PMC7753623 DOI: 10.3389/fimmu.2020.614972] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/04/2020] [Indexed: 12/12/2022] Open
Abstract
Human induced Pluripotent Stem Cell (hiPSC) models are a valuable new tool for research into neurodegenerative diseases. Neuroinflammation is now recognized as a key process in neurodegenerative disease and aging, and microglia are central players in this. A plethora of hiPSC-derived microglial models have been published recently to explore neuroinflammation, ranging from monoculture through to xenotransplantation. However, combining physiological relevance, reproducibility, and scalability into one model is still a challenge. We examine key features of the in vitro microglial environment, especially media composition, extracellular matrix, and co-culture, to identify areas for improvement in current hiPSC-microglia models.
Collapse
Affiliation(s)
| | | | | | - Sally A. Cowley
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
297
|
Spiteri AG, Wishart CL, King NJC. Immovable Object Meets Unstoppable Force? Dialogue Between Resident and Peripheral Myeloid Cells in the Inflamed Brain. Front Immunol 2020; 11:600822. [PMID: 33363542 PMCID: PMC7752943 DOI: 10.3389/fimmu.2020.600822] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammation of the brain parenchyma is characteristic of neurodegenerative, autoimmune, and neuroinflammatory diseases. During this process, microglia, which populate the embryonic brain and become a permanent sentinel myeloid population, are inexorably joined by peripherally derived monocytes, recruited by the central nervous system. These cells can quickly adopt a morphology and immunophenotype similar to microglia. Both microglia and monocytes have been implicated in inducing, enhancing, and/or maintaining immune-mediated pathology and thus disease progression in a number of neuropathologies. For many years, experimental and analytical systems have failed to differentiate resident microglia from peripherally derived myeloid cells accurately. This has impeded our understanding of their precise functions in, and contributions to, these diseases, and hampered the development of novel treatments that could target specific cell subsets. Over the past decade, microglia have been investigated more intensively in the context of neuroimmunological research, fostering the development of more precise experimental systems. In light of our rapidly growing understanding of these cells, we discuss the differential origins of microglia and peripherally derived myeloid cells in the inflamed brain, with an analysis of the problems resolving these cell types phenotypically and morphologically, and highlight recent developments enabling more precise identification.
Collapse
Affiliation(s)
- Alanna G. Spiteri
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Claire L. Wishart
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas J. C. King
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Cytometry Facility, The University of Sydney and Centenary Institute, Sydney, NSW, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Nano Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
298
|
Zhang J, Velmeshev D, Hashimoto K, Huang YH, Hofmann JW, Shi X, Chen J, Leidal AM, Dishart JG, Cahill MK, Kelley KW, Liddelow SA, Seeley WW, Miller BL, Walther TC, Farese RV, Taylor JP, Ullian EM, Huang B, Debnath J, Wittmann T, Kriegstein AR, Huang EJ. Neurotoxic microglia promote TDP-43 proteinopathy in progranulin deficiency. Nature 2020; 588:459-465. [PMID: 32866962 PMCID: PMC7746606 DOI: 10.1038/s41586-020-2709-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/21/2020] [Indexed: 12/21/2022]
Abstract
Aberrant aggregation of the RNA-binding protein TDP-43 in neurons is a hallmark of frontotemporal lobar degeneration caused by haploinsufficiency in the gene encoding progranulin1,2. However, the mechanism leading to TDP-43 proteinopathy remains unclear. Here we use single-nucleus RNA sequencing to show that progranulin deficiency promotes microglial transition from a homeostatic to a disease-specific state that causes endolysosomal dysfunction and neurodegeneration in mice. These defects persist even when Grn-/- microglia are cultured ex vivo. In addition, single-nucleus RNA sequencing reveals selective loss of excitatory neurons at disease end-stage, which is characterized by prominent nuclear and cytoplasmic TDP-43 granules and nuclear pore defects. Remarkably, conditioned media from Grn-/- microglia are sufficient to promote TDP-43 granule formation, nuclear pore defects and cell death in excitatory neurons via the complement activation pathway. Consistent with these results, deletion of the genes encoding C1qa and C3 mitigates microglial toxicity and rescues TDP-43 proteinopathy and neurodegeneration. These results uncover previously unappreciated contributions of chronic microglial toxicity to TDP-43 proteinopathy during neurodegeneration.
Collapse
Affiliation(s)
- Jiasheng Zhang
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Pathology Service 113B, San Francisco VA Health Care System, San Francisco, CA, USA
| | - Dmitry Velmeshev
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Kei Hashimoto
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Yu-Hsin Huang
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Jeffrey W Hofmann
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Xiaoyu Shi
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Jiapei Chen
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Andrew M Leidal
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Julian G Dishart
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Michelle K Cahill
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Kevin W Kelley
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Shane A Liddelow
- Neuroscience Institute, Department of Neuroscience & Physiology, NYU Langone Medical Center, New York, NY, USA
| | - William W Seeley
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Tobias C Walther
- Department of Genetics and Complex Diseases, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Robert V Farese
- Department of Genetics and Complex Diseases, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - J Paul Taylor
- Department of Cell and Molecular Biology, St Jude Children's Hospital & Howard Hughes Medical Institute, Memphis, TN, USA
| | - Erik M Ullian
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jayanta Debnath
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Torsten Wittmann
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - Arnold R Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Eric J Huang
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA.
- Pathology Service 113B, San Francisco VA Health Care System, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
299
|
Li L, Shi Y. When glia meet induced pluripotent stem cells (iPSCs). Mol Cell Neurosci 2020; 109:103565. [PMID: 33068719 PMCID: PMC10506562 DOI: 10.1016/j.mcn.2020.103565] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 09/20/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
The importance of glial cells, mainly astrocytes, oligodendrocytes, and microglia, in the central nervous system (CNS) has been increasingly appreciated. Recent advances have demonstrated the diversity of glial cells and their contribution to human CNS development, normal CNS functions, and disease progression. The uniqueness of human glial cells is also supported by multiple lines of evidence. With the discovery of induced pluripotent stem cells (iPSCs) and the progress of generating glial cells from human iPSCs, there are numerous studies to model CNS diseases using human iPSC-derived glial cells. Here we summarize the basic characteristics of glial cells, with the focus on their classical functions, heterogeneity, and uniqueness in human species. We further review the findings from recent studies that use iPSC-derived glial cells for CNS disease modeling. We conclude with promises and future directions of using iPSC-derived glial cells for CNS disease modeling.
Collapse
Affiliation(s)
- Li Li
- Division of Stem Cell Biology, Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Yanhong Shi
- Division of Stem Cell Biology, Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
300
|
Understanding and Treating Niemann-Pick Type C Disease: Models Matter. Int J Mol Sci 2020; 21:ijms21238979. [PMID: 33256121 PMCID: PMC7730076 DOI: 10.3390/ijms21238979] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023] Open
Abstract
Biomedical research aims to understand the molecular mechanisms causing human diseases and to develop curative therapies. So far, these goals have been achieved for a small fraction of diseases, limiting factors being the availability, validity, and use of experimental models. Niemann–Pick type C (NPC) is a prime example for a disease that lacks a curative therapy despite substantial breakthroughs. This rare, fatal, and autosomal-recessive disorder is caused by defects in NPC1 or NPC2. These ubiquitously expressed proteins help cholesterol exit from the endosomal–lysosomal system. The dysfunction of either causes an aberrant accumulation of lipids with patients presenting a large range of disease onset, neurovisceral symptoms, and life span. Here, we note general aspects of experimental models, we describe the line-up used for NPC-related research and therapy development, and we provide an outlook on future topics.
Collapse
|