1601
|
Shao QH, Zhang XL, Chen Y, Zhu CG, Shi JG, Yuan YH, Chen NH. Anti-neuroinflammatory effects of 20C from Gastrodia elata via regulating autophagy in LPS-activated BV-2 cells through MAPKs and TLR4/Akt/mTOR signaling pathways. Mol Immunol 2018; 99:115-123. [PMID: 29763880 DOI: 10.1016/j.molimm.2018.04.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/21/2018] [Accepted: 04/28/2018] [Indexed: 11/19/2022]
Abstract
20C, a novel bibenzyl compound, is isolated from Gastrodia elata. In our previous study, 20C showed protective effects on tunicamycin-induced endoplasmic reticulum stress, rotenone-induced apoptosis and rotenone-induced oxidative damage. However, the anti-neuroinflammatory effect of 20C is still with limited acquaintance. The objective of this study was to confirm the anti-neuroinflammatory effect of 20C on Lipopolysaccharide (LPS)-activated BV-2 cells and further elucidated the underlying molecular mechanisms. In this study, 20C significantly attenuated the protein levels of nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and interleukin (IL)-1β, and secretion of nitric oxide (NO) and tumor necrosis factor (TNF)-α induced by Lipopolysaccharide (LPS) in BV-2 cells. Moreover, 20C up-regulated the levels of autophagy-related proteins in LPS-activated BV-2 cells. The requirement of mitogen-activated protein kinases (MAPKs) has been well documented for regulating the process of autophagy. Both 20C and rapamycin enhanced autophagy by suppressing the phosphorylation of MAPKs signaling pathway. Furthermore, 20C treatment significantly inhibited the levels of toll like receptor 4 (TLR4), phosphorylated-protein kinase B (Akt) and phosphorylated-mechanistic target of rapamycin (mTOR), indicating blocking TLR4/Akt/mTOR might be an underlying basis for the anti-inflammatory effect of 20C. These findings suggest that 20C has therapeutic potential for treating neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Qian-Hang Shao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiao-Ling Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Cheng-Gen Zhu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jian-Gong Shi
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; College of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
1602
|
Abstract
Microglia are a subset of tissue macrophages that constitute the major immune cell type of the central nervous system. These cells have long been known to change their morphology and functions in response to various neurological insults. Recently, a plethora of unbiased transcriptomics studies have revealed that across a broad spectrum of neurodegeneration-like disease models, microglia adopt a similar activation signature and perform similar functions. Despite these commonalities in response, the role of microglia has been described as both positive and negative in different murine disease models. In humans, genetic association studies have revealed strong connections between microglia genes and various neurodegenerative diseases, and mechanistic investigations of these mutations have added another layer of complexity. Here, we provide an overview of studies that have built a case for a common microglial response to neurodegeneration and discuss pathways that may be important to initiate and sustain this response; delineate the multifaceted functions of activated microglia spanning different diseases; and discuss insights from studying genes associated with disease in humans. We argue that strong evidence causally links activated microglia function to neurodegeneration and discuss what seems to be a conflict between mouse models and human genetics.
Collapse
Affiliation(s)
- Wilbur M Song
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
1603
|
Cao K, Liao X, Lu J, Yao S, Wu F, Zhu X, Shi D, Wen S, Liu L, Zhou H. IL-33/ST2 plays a critical role in endothelial cell activation and microglia-mediated neuroinflammation modulation. J Neuroinflammation 2018; 15:136. [PMID: 29728120 PMCID: PMC5935936 DOI: 10.1186/s12974-018-1169-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 04/19/2018] [Indexed: 12/11/2022] Open
Abstract
Background Interleukin-33 (IL-33) is increasingly being recognized as a key immunomodulatory cytokine in many neurological diseases. Methods In the present study, wild-type (WT) and IL-33−/− mice received intracerebroventricular (i.c.v.) injection of lipopolysaccharide (LPS) to induce neuroinflammation. Intravital microscopy was employed to examine leukocyte–endothelial interactions in the brain vasculature. The degree of neutrophil infiltration was determined by myeloperoxidase (MPO) staining. Real-time PCR and western blotting were used to detect endothelial activation. Enzyme-linked immunosorbent assay and quantitative PCR were conducted to detect pro-inflammatory cytokine levels in the brain. Results In IL-33−/− mice, neutrophil infiltration in the brain cortex and leukocyte–endothelial cell interactions in the cerebral microvessels were significantly decreased as compared to WT mice after LPS injection. In addition, IL-33−/− mice showed reduced activation of microglia and cerebral endothelial cells. In vitro results indicated that IL-33 directly activated cerebral endothelial cells and promoted pro-inflammatory cytokine production in LPS-stimulated microglia. Conclusions Our study indicated that IL-33/ST2 signaling plays an important role in the activation of microglia and cerebral endothelial cells and, therefore, is essential in leukocyte recruitment in brain inflammation. Graphical abstract The role of IL-33/ST2 in LPS induced neuroinflammation![]()
Collapse
Affiliation(s)
- Kelei Cao
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Xiang Liao
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Jiahui Lu
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Shu Yao
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Fengjiao Wu
- Department of Immunology, Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, 233030, China
| | - Xingxing Zhu
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Dongyan Shi
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Shuang Wen
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Lixin Liu
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5, Canada
| | - Hong Zhou
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China.
| |
Collapse
|
1604
|
Losada-Perez M. Glia: from 'just glue' to essential players in complex nervous systems: a comparative view from flies to mammals. J Neurogenet 2018; 32:78-91. [PMID: 29718753 DOI: 10.1080/01677063.2018.1464568] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In the last years, glial cells have emerged as central players in the development and function of complex nervous systems. Therefore, the concept of glial cells has evolved from simple supporting cells to essential actors. The molecular mechanisms that govern glial functions are evolutionarily conserved from Drosophila to mammals, highlighting genetic similarities between these groups, as well as the great potential of Drosophila research for the understanding of human CNS. These similarities would imply a common phylogenetic origin of glia, even though there is a controversy at this point. This review addresses the existing literature on the evolutionary origin of glia and discusses whether or not insect and mammalian glia are homologous or analogous. Besides, this manuscript summarizes the main glial functions in the CNS and underscores the evolutionarily conserved molecular mechanisms between Drosophila and mammals. Finally, I also consider the current nomenclature and classification of glial cells to highlight the need for a consensus agreement and I propose an alternative nomenclature based on function that unifies Drosophila and mammalian glial types.
Collapse
|
1605
|
Macrophage Depletion Ameliorates Peripheral Neuropathy in Aging Mice. J Neurosci 2018; 38:4610-4620. [PMID: 29712789 DOI: 10.1523/jneurosci.3030-17.2018] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 02/06/2018] [Accepted: 03/09/2018] [Indexed: 01/02/2023] Open
Abstract
Aging is known as a major risk factor for the structure and function of the nervous system. There is urgent need to overcome such deleterious effects of age-related neurodegeneration. Here we show that peripheral nerves of 24-month-old aging C57BL/6 mice of either sex show similar pathological alterations as nerves from aging human individuals, whereas 12-month-old adult mice lack such alterations. Specifically, nerve fibers showed demyelination, remyelination and axonal lesion. Moreover, in the aging mice, neuromuscular junctions showed features typical for dying-back neuropathies, as revealed by a decline of presynaptic markers, associated with α-bungarotoxin-positive postsynapses. In line with these observations were reduced muscle strengths. These alterations were accompanied by elevated numbers of endoneurial macrophages, partially comprising the features of phagocytosing macrophages. Comparable profiles of macrophages could be identified in peripheral nerve biopsies of aging persons. To determine the pathological impact of macrophages in aging mice, we selectively targeted the cells by applying an orally administered CSF-1R specific kinase (c-FMS) inhibitor. The 6-month-lasting treatment started before development of degenerative changes at 18 months and reduced macrophage numbers in mice by ∼70%, without side effects. Strikingly, nerve structure was ameliorated and muscle strength preserved. We show, for the first time, that age-related degenerative changes in peripheral nerves are driven by macrophages. These findings may pave the way for treating degeneration in the aging peripheral nervous system by targeting macrophages, leading to reduced weakness, improved mobility, and eventually increased quality of life in the elderly.SIGNIFICANCE STATEMENT Aging is a major risk factor for the structure and function of the nervous system. Here we show that peripheral nerves of 24-month-old aging mice show similar degenerative alterations as nerves from aging human individuals. Both in mice and humans, these alterations were accompanied by endoneurial macrophages. To determine the pathological impact of macrophages in aging mice, we selectively targeted the cells by blocking a cytokine receptor, essential for macrophage survival. The treatment strongly reduced macrophage numbers and substantially improved nerve structure and muscle strength. We show, for the first time, that age-related degenerative changes in peripheral nerves are driven by macrophages. These findings may be helpful for treatment weakness and reduced mobility in the elderly.
Collapse
|
1606
|
The role of catecholamines in HIV neuropathogenesis. Brain Res 2018; 1702:54-73. [PMID: 29705605 DOI: 10.1016/j.brainres.2018.04.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/21/2018] [Accepted: 04/24/2018] [Indexed: 12/21/2022]
Abstract
The success of anti-retroviral therapy has improved the quality of life and lifespan of HIV + individuals, transforming HIV infection into a chronic condition. These improvements have come with a cost, as chronic HIV infection and long-term therapy have resulted in the emergence of a number of new pathologies. This includes a variety of the neuropathological and neurocognitive effects collectively known as HIVassociated neurocognitive disorders (HAND) or NeuroHIV. These effects persist even in the absence of viral replication, suggesting that they are mediated the long-term changes in the CNS induced by HIV infection rather than by active replication. Among these effects are significant changes in catecholaminergic neurotransmission, especially in dopaminergic brain regions. In HIV-infected individuals not treated with ARV show prominent neuropathology is common in dopamine-rich brain regions and altered autonomic nervous system activity. Even infected individuals on therapy, there is significant dopaminergic neuropathology, and elevated stress and norepinephrine levels correlate with a decreased effectiveness of antiretroviral drugs. As catecholamines function as immunomodulatory factors, the resultant dysregulation of catecholaminergic tone could substantially alter the development of HIVassociated neuroinflammation and neuropathology. In this review, we discuss the role of catecholamines in the etiology of HIV neuropathogenesis. Providing a comprehensive examination of what is known about these molecules in the context of HIV-associated disease demonstrates the importance of further studies in this area, and may open the door to new therapeutic strategies that specifically ameliorate the effects of catecholaminergic dysregulation on NeuroHIV.
Collapse
|
1607
|
Lee SH, Suk K. Identification of glia phenotype modulators based on select glial function regulatory signaling pathways. Expert Opin Drug Discov 2018; 13:627-641. [DOI: 10.1080/17460441.2018.1465925] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Sun-Hwa Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science & Engineering Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
1608
|
Heindl S, Gesierich B, Benakis C, Llovera G, Duering M, Liesz A. Automated Morphological Analysis of Microglia After Stroke. Front Cell Neurosci 2018; 12:106. [PMID: 29725290 PMCID: PMC5917008 DOI: 10.3389/fncel.2018.00106] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/03/2018] [Indexed: 01/10/2023] Open
Abstract
Microglia are the resident immune cells of the brain and react quickly to changes in their environment with transcriptional regulation and morphological changes. Brain tissue injury such as ischemic stroke induces a local inflammatory response encompassing microglial activation. The change in activation status of a microglia is reflected in its gradual morphological transformation from a highly ramified into a less ramified or amoeboid cell shape. For this reason, the morphological changes of microglia are widely utilized to quantify microglial activation and studying their involvement in virtually all brain diseases. However, the currently available methods, which are mainly based on manual rating of immunofluorescent microscopic images, are often inaccurate, rater biased, and highly time consuming. To address these issues, we created a fully automated image analysis tool, which enables the analysis of microglia morphology from a confocal Z-stack and providing up to 59 morphological features. We developed the algorithm on an exploratory dataset of microglial cells from a stroke mouse model and validated the findings on an independent data set. In both datasets, we could demonstrate the ability of the algorithm to sensitively discriminate between the microglia morphology in the peri-infarct and the contralateral, unaffected cortex. Dimensionality reduction by principal component analysis allowed to generate a highly sensitive compound score for microglial shape analysis. Finally, we tested for concordance of results between the novel automated analysis tool and the conventional manual analysis and found a high degree of correlation. In conclusion, our novel method for the fully automatized analysis of microglia morphology shows excellent accuracy and time efficacy compared to traditional analysis methods. This tool, which we make openly available, could find application to study microglia morphology using fluorescence imaging in a wide range of brain disease models.
Collapse
Affiliation(s)
- Steffanie Heindl
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Benno Gesierich
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Corinne Benakis
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gemma Llovera
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
1609
|
Paolicelli RC, Bergamini G, Rajendran L. Cell-to-cell Communication by Extracellular Vesicles: Focus on Microglia. Neuroscience 2018; 405:148-157. [PMID: 29660443 DOI: 10.1016/j.neuroscience.2018.04.003] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/30/2018] [Accepted: 04/04/2018] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles, including exosomes and microvesicles, are small, nano-to-micrometer vesicles that are released from cells. While initially observed in immune cells and reticulocytes as vesicles meant to remove archaic proteins, now they have been observed in almost all cell types of multicellular organisms. Growing evidence indicates that extracellular vesicles, containing lipids, proteins and RNAs, represent an efficient way to transfer functional cargoes from one cell to another. In the central nervous system, the extensive cross-talk ongoing between neurons and glia, including microglia, the immune cells of the brain, takes advantage of secreted vesicles, which mediate intercellular communication over long range distance. Recent literature supports a critical role for extracellular vesicles in mediating complex and coordinated communication among neurons, astrocytes and microglia, both in the healthy and in the diseased brain. In this review, we focus on the biogenesis and function of microglia-related extracellular vesicles and focus on their putative role in Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Rosa C Paolicelli
- Systems and Cell Biology of Neurodegeneration, IREM - Institute for Regenerative Medicine, University of Zurich, Switzerland.
| | - Giorgio Bergamini
- Preclinical Laboratory for Translational Research into Affective Disorders (PLaTRAD), Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Switzerland
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, IREM - Institute for Regenerative Medicine, University of Zurich, Switzerland
| |
Collapse
|
1610
|
You MM, Chen YF, Pan YM, Liu YC, Tu J, Wang K, Hu FL. Royal Jelly Attenuates LPS-Induced Inflammation in BV-2 Microglial Cells through Modulating NF- κB and p38/JNK Signaling Pathways. Mediators Inflamm 2018; 2018:7834381. [PMID: 29849495 PMCID: PMC5911318 DOI: 10.1155/2018/7834381] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/05/2018] [Accepted: 03/08/2018] [Indexed: 12/13/2022] Open
Abstract
Royal jelly (RJ), a hive product with versatile pharmacological activities, has been used as a traditional functional food to prevent or treat inflammatory diseases. However, little is known about the anti-inflammatory effect of RJ in microglial cells. The aim of this study is to assess the anti-inflammatory effects of RJ in lipopolysaccharide- (LPS-) induced murine immortalized BV-2 cells and to explore the underlying molecular mechanisms. Our results showed that in LPS-stimulated BV-2 cells, RJ significantly inhibited iNOS and COX-2 expression at mRNA and protein levels. The mRNA expression of IL-6, IL-1β, and TNF-α was also downregulated by RJ in a concentration-dependent manner. Additionally, RJ protected BV-2 cells against oxidative stress by upregulating heme oxygenase-1 (HO-1) expression and by reducing reactive oxygen species (ROS) and nitric oxide (NO) production. Mechanistically, we found that RJ could alleviate inflammatory response in microglia by suppressing the phosphorylation of IκBα, p38, and JNK and by inhibiting the nucleus translocation of NF-κB p65. These findings suggest that RJ might be a promising functional food to delay inflammatory progress by influencing the microglia function.
Collapse
Affiliation(s)
- Meng-Meng You
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi-Fan Chen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yong-Ming Pan
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Comparative Medical Research Center, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi-Chen Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jue Tu
- Comparative Medical Research Center, Experimental Animal Research Center, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Kai Wang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Fu-Liang Hu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
1611
|
Kim HY, Spector AA. N-Docosahexaenoylethanolamine: A neurotrophic and neuroprotective metabolite of docosahexaenoic acid. Mol Aspects Med 2018; 64:34-44. [PMID: 29572109 DOI: 10.1016/j.mam.2018.03.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 12/11/2022]
Abstract
N-Docosahexaenoylethanolamine (synaptamide) is an endocannabinoid-like metabolite endogenously synthesized from docosahexaenoic acid (DHA, 22:6n-3), the major omega-3 polyunsaturated fatty acid present in the brain. Although its biosynthetic mechanism has yet to be established, there is a closely linked relationship between the levels of synaptamide and its precursor DHA in the brain. Synaptamide at nanomolar concentrations promotes neurogenesis, neurite outgrowth and synaptogenesis in developing neurons. Synaptamide also attenuates the lipopolysaccharide-induced neuroinflammatory response and reduces the deleterious effects of ethanol on neurogenic differentiation of neural stem cells (NSCs). These actions are mediated by a specific target receptor of synaptamide GPR110 (ADGRF1), a G-protein coupled receptor that is highly expressed in NSCs and the brain during development. Synaptamide binding to GPR110 induces cAMP production and phosphorylation of protein kinase A (PKA) and the cAMP response element binding protein (CREB). This signaling pathway leads to the expression of neurogenic and synaptogenic genes and suppresses the expression of proinflammatory genes. The GPR110-dependent cellular effects of synaptamide are recapitulated in animal models, suggesting that synaptamide-derived mechanisms may have translational implications. The synaptamide bioactivity transmitted by newly deorphanized GPR110 provides a novel target for neurodevelopmental and neuroprotective control as well as new insight into mechanisms for DHA's beneficial effects on the central nervous system.
Collapse
Affiliation(s)
- Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-9410, United States.
| | - Arthur A Spector
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-9410, United States
| |
Collapse
|
1612
|
Histone Deacetylases 1 and 2 Regulate Microglia Function during Development, Homeostasis, and Neurodegeneration in a Context-Dependent Manner. Immunity 2018; 48:514-529.e6. [PMID: 29548672 DOI: 10.1016/j.immuni.2018.02.016] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 12/21/2017] [Accepted: 02/22/2018] [Indexed: 11/20/2022]
Abstract
Microglia as tissue macrophages contribute to the defense and maintenance of central nervous system (CNS) homeostasis. Little is known about the epigenetic signals controlling microglia function in vivo. We employed constitutive and inducible mutagenesis in microglia to delete two class I histone deacetylases, Hdac1 and Hdac2. Prenatal ablation of Hdac1 and Hdac2 impaired microglial development. Mechanistically, the promoters of pro-apoptotic and cell cycle genes were hyperacetylated in absence of Hdac1 and Hdac2, leading to increased apoptosis and reduced survival. In contrast, Hdac1 and Hdac2 were not required for adult microglia survival during homeostasis. In a mouse model of Alzheimer's disease, deletion of Hdac1 and Hdac2 in microglia, but not in neuroectodermal cells, resulted in a decrease in amyloid load and improved cognitive impairment by enhancing microglial amyloid phagocytosis. Collectively, we report a role for epigenetic factors that differentially affect microglia development, homeostasis, and disease that could potentially be utilized therapeutically.
Collapse
|
1613
|
Collins HY, Bohlen CJ. Isolation and Culture of Rodent Microglia to Promote a Dynamic Ramified Morphology in Serum-free Medium. J Vis Exp 2018. [PMID: 29578519 DOI: 10.3791/57122] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Microglia represent 5 - 10% of all central nervous system (CNS) cells and are increasingly drawing attention due to their contributions during development, homeostasis, and disease. Although macrophages have been studied in detail for decades, specialized features of microglia, the tissue-resident macrophages of the CNS, have remained largely mysterious, in part due to limitations in the ability to recapitulate mature microglial properties in culture. Here, we illustrate a straightforward procedure for the rapid isolation of pure microglia from the mature rodent brain. We also describe serum-free culture conditions that support high levels of microglial viability over time. Microglia cultured under these defined-medium conditions exhibit elaborate ramified processes and dynamic surveillance behavior. We illustrate some effects of serum exposure on cultured microglia and discuss how these serum-free cultures compare to both serum-exposed cultures as well as microglia in vivo.
Collapse
|
1614
|
Abstract
Neuroinflammation, which involves microglial activation, is thought to play a key role in the development and progression of neurodegenerative diseases and other brain pathologies. Positron emission tomography is an ideal imaging technique for studying biochemical processes in vivo, and particularly for studying the living brain. Neuroinflammation has been traditionally studied using radiotracers targeting the translocator protein 18 kDa, but this comes with certain limitations. The current review describes alternative biological targets that have gained interest for the imaging of microglial activation over recent years, such as the cannabinoid receptor type 2, cyclooxygenase-2, the P2X₇ receptor and reactive oxygen species, and some promising radiotracers for these targets. Although many advances have been made in the field of neuroinflammation imaging, current radiotracers all target the pro-inflammatory (M1) phenotype of activated microglia, since the number of known biological targets specific for the anti-inflammatory (M2) phenotype that are also suited as a target for radiotracer development is still limited. Next to proceeding the currently available tracers for M1 microglia into the clinic, the development of a suitable radiotracer for M2 microglia would mean a great advance in the field, as this would allow for imaging of the dynamics of microglial activation in different diseases.
Collapse
Affiliation(s)
- Bieneke Janssen
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Danielle J Vugts
- Department of Radiology & Nuclear Medicine, VU University Medical Center, 1081 HV Amsterdam, The Netherlands.
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, 1081 HV Amsterdam, The Netherlands.
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
1615
|
Calvani R, Picca A, Lo Monaco MR, Landi F, Bernabei R, Marzetti E. Of Microbes and Minds: A Narrative Review on the Second Brain Aging. Front Med (Lausanne) 2018; 5:53. [PMID: 29552561 PMCID: PMC5840854 DOI: 10.3389/fmed.2018.00053] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 02/15/2018] [Indexed: 12/21/2022] Open
Abstract
In recent years, an extensive body of literature focused on the gut-brain axis and the possible role played by the gut microbiota in modulating brain morphology and function from birth to old age. Gut microbiota has been proposed as a relevant player during the early phases of neurodevelopment, with possible long-standing effects in later life. The reduction in gut microbiota diversity has also become one of the hallmarks of aging, and disturbances in its composition are associated with several (age-related) neurological conditions, including depression, Alzheimer's disease, and Parkinson's disease. Several pathways have been evoked for gut microbiota-brain communication, including neural connections (vagus nerve), circulating mediators derived by host-bacteria cometabolism, as well as the influence exerted by gut microbiota on host gut function, metabolism, and immune system. Although the most provoking data emerged from animal studies and despite the huge debate around the possible epiphenomenal nature of those findings, the gut microbiota-brain axis still remains a fascinating target to be exploited to attenuate some of the most burdensome consequences of aging.
Collapse
Affiliation(s)
- Riccardo Calvani
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Anna Picca
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Maria Rita Lo Monaco
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco Landi
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Roberto Bernabei
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
1616
|
Chelini G, Pantazopoulos H, Durning P, Berretta S. The tetrapartite synapse: a key concept in the pathophysiology of schizophrenia. Eur Psychiatry 2018; 50:60-69. [PMID: 29503098 PMCID: PMC5963512 DOI: 10.1016/j.eurpsy.2018.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 02/01/2018] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
Growing evidence points to synaptic pathology as a core component of the pathophysiology of schizophrenia (SZ). Significant reductions of dendritic spine density and altered expression of their structural and molecular components have been reported in several brain regions, suggesting a deficit of synaptic plasticity. Regulation of synaptic plasticity is a complex process, one that requires not only interactions between pre- and post-synaptic terminals, but also glial cells and the extracellular matrix (ECM). Together, these elements are referred to as the ‘tetrapartite synapse’, an emerging concept supported by accumulating evidence for a role of glial cells and the extracellular matrix in regulating structural and functional aspects of synaptic plasticity. In particular, chondroitin sulfate proteoglycans (CSPGs), one of the main components of the ECM, have been shown to be synthesized predominantly by glial cells, to form organized perisynaptic aggregates known as perineuronal nets (PNNs), and to modulate synaptic signaling and plasticity during postnatal development and adulthood. Notably, recent findings from our group and others have shown marked CSPG abnormalities in several brain regions of people with SZ. These abnormalities were found to affect specialized ECM structures, including PNNs, as well as glial cells expressing the corresponding CSPGs. The purpose of this review is to bring forth the hypothesis that synaptic pathology in SZ arises from a disruption of the interactions between elements of the tetrapartite synapse.
Collapse
Affiliation(s)
- Gabriele Chelini
- Translational Neuroscience Laboratory, Mclean Hospital, 115 Mill Street, Belmont, MA, 02478 USA; Dept. of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115 USA.
| | - Harry Pantazopoulos
- Translational Neuroscience Laboratory, Mclean Hospital, 115 Mill Street, Belmont, MA, 02478 USA; Dept. of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115 USA.
| | - Peter Durning
- Translational Neuroscience Laboratory, Mclean Hospital, 115 Mill Street, Belmont, MA, 02478 USA.
| | - Sabina Berretta
- Translational Neuroscience Laboratory, Mclean Hospital, 115 Mill Street, Belmont, MA, 02478 USA; Dept. of Psychiatry, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115 USA; Program in Neuroscience, Harvard Medical School, 220 Longwood Ave., Boston, MA, 02115 USA.
| |
Collapse
|
1617
|
Zhang Y, Zhao L, Wang X, Ma W, Lazere A, Qian HH, Zhang J, Abu-Asab M, Fariss RN, Roger JE, Wong WT. Repopulating retinal microglia restore endogenous organization and function under CX3CL1-CX3CR1 regulation. SCIENCE ADVANCES 2018; 4:eaap8492. [PMID: 29750189 PMCID: PMC5943055 DOI: 10.1126/sciadv.aap8492] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/12/2018] [Indexed: 05/30/2023]
Abstract
Microglia have been discovered to undergo repopulation following ablation. However, the functionality of repopulated microglia and the mechanisms regulating microglia repopulation are unknown. We examined microglial homeostasis in the adult mouse retina, a specialized neural compartment containing regular arrays of microglia in discrete synaptic laminae that can be directly visualized. Using in vivo imaging and cell-fate mapping techniques, we discovered that repopulation originated from residual microglia proliferating in the central inner retina that subsequently spread by centrifugal migration to fully recapitulate pre-existing microglial distributions and morphologies. Repopulating cells fully restored microglial functions including constitutive "surveying" process movements, behavioral and physiological responses to retinal injury, and maintenance of synaptic structure and function. Microglial repopulation was regulated by CX3CL1-CX3CR1 signaling, slowing in CX3CR1 deficiency and accelerating with exogenous CX3CL1 administration. Microglial homeostasis following perturbation can fully recover microglial organization and function under the regulation of chemokine signaling between neurons and microglia.
Collapse
Affiliation(s)
- Yikui Zhang
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Lian Zhao
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xu Wang
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenxin Ma
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adam Lazere
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hao-hua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun Zhang
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mones Abu-Asab
- Section on Histopathology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert N. Fariss
- Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jerome E. Roger
- Centre d’Etude et de Recherche Thérapeutique en Ophtalmologie (CERTO), Retina France, Orsay, France
- Paris-Saclay Institute of Neuroscience, CNRS, Université Paris-Sud, Université Paris-Saclay, Orsay, France
| | - Wai T. Wong
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
1618
|
Frank MG, Fonken LK, Annis JL, Watkins LR, Maier SF. Stress disinhibits microglia via down-regulation of CD200R: A mechanism of neuroinflammatory priming. Brain Behav Immun 2018; 69:62-73. [PMID: 29104062 PMCID: PMC5857401 DOI: 10.1016/j.bbi.2017.11.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/20/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022] Open
Abstract
Exposure to stressors primes the neuroinflammatory and microglial proinflammatory response to subsequent immune challenges, suggesting that stress might attenuate immunoregulatory mechanisms in the CNS microenvironment. CD200:CD200R is a key immunoregulatory signaling dyad that constrains microglial activation, and disruption of CD200:CD200R signaling primes microglia to subsequent immune challenges. Therefore, the present study examined the mediating role of CD200:CD200R signaling in stress-induced microglial priming. Here, we found that exposure to an acute stressor reduced CD200R expression across sub-regions of the hippocampus, amygdala as well as in isolated hippocampal microglia. A transcriptional suppressor of CD200R, CAAT/Enhancer Binding Proteinβ, was induced by stress and inversely associated with CD200R expression. To examine whether disrupted CD200:CD200R signaling plays a mediating role in stress-induced microglial priming, a soluble fragment of CD200 (mCD200Fc) was administered intra-cisterna magna prior to stressor exposure and stress-induced microglia priming assessed ex vivo 24 h later. Treatment with mCD200Fc blocked the stress-induced priming of the microglial pro-inflammatory response. Further, treatment with mCD200R1Fc recapitulated the effects of stress on microglial priming. We previously found that stress increases the alarmin high mobility group box-1 (HMGB1) in hippocampus, and that HMGB1 mediates stress-induced priming of microglia. Thus, we examined whether stress-induced increases in hippocampal HMGB1 are a consequence of disrupted CD200:CD200R signaling. Indeed, treatment with mCD200Fc prior to stress exposure blocked the stress-induced increase in hippocampal HMGB1. The present study suggests that stress exposure disrupts immunoregulatory mechanisms in the brain, which typically constrain the immune response of CNS innate immune cells. This attenuation of immunoregulatory mechanisms may thus permit a primed activation state of microglia to manifest.
Collapse
Affiliation(s)
- Matthew G. Frank
- Corresponding Author: Department of Psychology and Neuroscience, Center for Neuroscience, Campus Box 345, University of Colorado Boulder, Boulder, CO, 80309-0345, USA, Tel: +1-303-919-8116, Fax: +1-303-492-2967,
| | | | | | | | | |
Collapse
|
1619
|
Franceschini A, Strammiello R, Capellari S, Giese A, Parchi P. Regional pattern of microgliosis in sporadic Creutzfeldt-Jakob disease in relation to phenotypic variants and disease progression. Neuropathol Appl Neurobiol 2018; 44:574-589. [PMID: 29345730 DOI: 10.1111/nan.12461] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 12/22/2017] [Indexed: 12/30/2022]
Abstract
AIMS The aim of this study was to describe the regional profiles of microglial activation in sporadic Creutzfeldt-Jakob disease (sCJD) subtypes and analyse the influence of prion strain, disease duration and codon 129 genotype. METHODS We studied the amount/severity and distribution of activated microglia, protease-resistant prion protein (PrPSc ) spongiform change, and astrogliosis in eight regions of 57 brains, representative of the entire spectrum of sCJD subtypes. RESULTS In each individual subtype, the regional extent and distribution of microgliosis significantly correlated with PrPSc deposition and spongiform change, leading to subtype-specific 'lesion profiles'. However, large differences in the ratio between PrPSc load or the score of spongiform change and microglial activation were seen among disease subtypes. Most significantly, atypical sCJD subtypes such as VV1 and MM2T showed a degree of microglial activation comparable to other disease variants despite the relatively low PrPSc deposition and the less severe spongiform change. Moreover, the mean microglial total load was significantly higher in subtype MM1 than in MM2C, whereas the opposite was true for the PrPSc and spongiform change total loads. Finally, some sCJD subtypes showed distinctive regional cerebellar profiles of microgliosis characterized by a high granular/molecular layer ratio (MV2K) and/or a predominant involvement of white matter (MVK and MM2T). CONCLUSIONS Microglial activation is an early event in sCJD pathogenesis and is strongly influenced by prion strain, PRNP codon 129 genotype and disease duration. Microglial lesion profiling, by highlighting strain-specific properties of prions, contributes to prion strain characterization and classification of human prion diseases, and represents a valid support to molecular and histopathologic typing.
Collapse
Affiliation(s)
- A Franceschini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - R Strammiello
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - S Capellari
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS, Institute of Neurological Sciences, Bologna, Italy
| | - A Giese
- Institut für Neuropathologie und Prion Forschung, Ludwig-Maximilians-Universität München, Munich, Germany
| | - P Parchi
- IRCCS, Institute of Neurological Sciences, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| |
Collapse
|
1620
|
Diffusion Basis Spectral Imaging Detects Ongoing Brain Inflammation in Virologically Well-Controlled HIV+ Patients. J Acquir Immune Defic Syndr 2018; 76:423-430. [PMID: 28796748 DOI: 10.1097/qai.0000000000001513] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Inflammation occurs after HIV infection and persists, despite highly active antiretroviral therapy (HAART). Diffusion tensor imaging (DTI) measures HIV-associated white matter changes, but can be confounded by inflammation. Currently, the influence of inflammation on white matter integrity in well-controlled HIV+ patients remains unknown. We used diffusion basis spectral imaging (DBSI)-derived cellularity to isolate restricted water diffusion associated with inflammation separated from the anisotropic diffusion associated with axonal integrity. Ninety-two virologically suppressed HIV+ patients on HAART and 66 HIV uninfected (HIV-) controls underwent neuropsychological performance (NP) testing and neuroimaging. NP tests assessed multiple domains (memory, psychomotor speed, and executive functioning). DTI- and DBSI-derived fractional anisotropy (FA) maps were processed with tract-based spatial statistics for comparison between both groups. Cellularity was assessed regarding age, HIV status, and NP. Within the HIV+ cohort, cellularity was compared with clinical (HAART duration) and laboratory measures of disease (eg, CD4 cell current and nadir). NP was similar for both groups. DTI-derived FA was lower in HIV+ compared with HIV- individuals. By contrast, DBSI-derived FA was similar for both groups. Instead, diffuse increases in cellularity were present in HIV+ individuals. Observed changes in cellularity were significantly associated with age, but not NP, in HIV+ individuals. A trend level association was seen between cellularity and HAART duration. Elevated inflammation, measured by cellularity, persists in virologically well-controlled HIV+ individuals. Widespread cellularity changes occur in younger HIV+ individuals and diminish with aging and duration of HAART.
Collapse
|
1621
|
Murgoci AN, Cizkova D, Majerova P, Petrovova E, Medvecky L, Fournier I, Salzet M. Brain-Cortex Microglia-Derived Exosomes: Nanoparticles for Glioma Therapy. Chemphyschem 2018; 19:1205-1214. [PMID: 29327816 DOI: 10.1002/cphc.201701198] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/03/2018] [Indexed: 12/19/2022]
Abstract
The function and integrity of the nervous system require interactive exchanges among neurons and glial cells. Exosomes and other extracellular vesicles (EVs) are emerging as a key mediator of intercellular communication, capable of transferring nucleic acids, proteins and lipids influencing numerous functional and pathological aspects of both donor and recipient cells. The immune response mediated by microglia-derived exosomes is most prominently involved in the spread of neuroinflammation, neurodegenerative disorders, and brain cancer. Therefore, in the present study we describe a reproducible and highly efficient method for yielding purified primary microglia cells, followed by exosome isolation and their characterization. An in vitro biological assay demonstrates that microglia-derived exosomes tested on a 3D spheroid glioma culture were able to inhibit tumor invasion in time course. These results evidence that brain microglia-derived exosomes could be used as nanotherapeutic agents against glioma cells.
Collapse
Affiliation(s)
- Adriana-Natalia Murgoci
- Univ. Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000, Lille, France.,Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10, Bratislava, Slovakia.,Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81, Košice, Slovakia
| | - Dasa Cizkova
- Univ. Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000, Lille, France.,Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10, Bratislava, Slovakia.,Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81, Košice, Slovakia
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10, Bratislava, Slovakia
| | - Eva Petrovova
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81, Košice, Slovakia
| | - Lubomir Medvecky
- Institute of Materials Research, Slovak Academy of Sciences, Watsonova 47, 040 01, Košice, Slovakia
| | - Isabelle Fournier
- Univ. Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000, Lille, France
| | - Michel Salzet
- Univ. Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000, Lille, France
| |
Collapse
|
1622
|
Plasma Hemopexin ameliorates murine spinal cord injury by switching microglia from the M1 state to the M2 state. Cell Death Dis 2018; 9:181. [PMID: 29415995 PMCID: PMC5833847 DOI: 10.1038/s41419-017-0236-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/28/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022]
Abstract
Spinal cord injury (SCI) is a devastating type of central nervous system (CNS) trauma with limited therapeutic treatments. The polarization of microglia into the M1 or M2 state has been documented to play important roles in the pathogenesis of SCI, although the complete repertoire of underlying factors has not been identified. Interestingly, the time point at which hematomyelia (intramedullary spinal cord hemorrhage) is alleviated coincides with a decrease in the number of M2 microglia. Here the function of Hemopexin (Hpx), a hematogenous glycoprotein, was examined in the crush model of SCI. Hpx levels were elevated at the lesion site during hematomyelia and were synchronously correlated with the level of the M2 marker Arginase-1 (Arg-1). Ablation of Hpx in vivo affected the polarization state of lipopolysaccharide (LPS)-stimulated microglia, as mirrored by a lower percentage of M2 microglia and a higher percentage of M1 microglia in the lesion site, which delayed the recovery and exacerbated the behavioral dysfunction after SCI. However, Hpx induced a rapid switch from the M1 to M2 phenotype in LPS-stimulated primary cultured microglia in a heme scavenging-independent manner. The supernant of Hpx-treated microglia ameliorated neuronal degeneration, alleviated demyelination, and promoted oligodendrocyte precursor cell (OPC) maturation. This modulatory effect of Hpx on microglia polarization was at least partially mediated by the LRP-1 receptor. Based on these results, Hpx is considered a novel modulator of the polarization of microglia during the pathogenesis of SCI and may play a crucial role in the recovery from SCI.
Collapse
|
1623
|
Rizzi N, Villa A, Benedusi V, Brunialti E, Cesari N, Ciana P, Maggi A. Endocrine influence on neuroinflammation: the use of reporter systems. J Neuroendocrinol 2018. [PMID: 28650102 DOI: 10.1111/jne.12496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Most of the ageing-associated pathologies are coupled with a strong inflammatory component that accelerates the progress of the physiopathological functional decline related to ageing. The currently available pharmacological tools for the control of neuroinflammation present several side effects that restrict their application, particularly in chronic disorders. The discovery of the potential anti-inflammatory action exerted by endogenous oestrogens, as well as the finding that activation of oestrogen receptor α results in a significant decrease of inflammation at the cellular level and in models of inflammatory diseases, prompted us to embark in a series of studies aimed at the generation of reporter systems, allowing us to (i) understand the anti-inflammatory action of oestrogens at molecular level; (ii) evaluate the extent to which the action of this steroid hormone was relevant in models of pathologies characterised by a strong inflammatory component; and (iii) investigate the efficacy of novel, synthetic oestrogens endowed with anti-inflammatory activity. Accordingly, we conceived the NFκB-luc2 reporter mouse, a model characterised by dual reporter genes for fluorescence and bioluminescence imaging under the control of a synthetic DNA able to bind the transcription factor nuclear factor kappa B, the master regulator of the expression of most of the cytokines responsible for the initial phase of acute inflammation. Here, we summarise the philosophy that has driven our research in the past years, as well as some of the results obtained so far.
Collapse
Affiliation(s)
- N Rizzi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence for Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - A Villa
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence for Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - V Benedusi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence for Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - E Brunialti
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence for Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - N Cesari
- Centro Clinico-Veterinario e Zootecnico-Sperimentale d'Ateneo, University of Milan, Lodi, Italy
| | - P Ciana
- Department of Oncology and Hemato-Oncology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - A Maggi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence for Neurodegenerative Diseases, University of Milan, Milan, Italy
| |
Collapse
|
1624
|
Mrdjen D, Pavlovic A, Hartmann FJ, Schreiner B, Utz SG, Leung BP, Lelios I, Heppner FL, Kipnis J, Merkler D, Greter M, Becher B. High-Dimensional Single-Cell Mapping of Central Nervous System Immune Cells Reveals Distinct Myeloid Subsets in Health, Aging, and Disease. Immunity 2018; 48:380-395.e6. [DOI: 10.1016/j.immuni.2018.01.011] [Citation(s) in RCA: 311] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/05/2017] [Accepted: 01/14/2018] [Indexed: 12/14/2022]
|
1625
|
Kim G, Vahedi S, Gefen T, Weintraub S, Bigio EH, Mesulam MM, Geula C. Asymmetric TDP pathology in primary progressive aphasia with right hemisphere language dominance. Neurology 2018; 90:e396-e403. [PMID: 29305438 PMCID: PMC5791793 DOI: 10.1212/wnl.0000000000004891] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/12/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To quantitatively examine the regional densities and hemispheric distribution of the 43-kDa transactive response DNA-binding protein (TDP-43) inclusions, neurons, and activated microglia in a left-handed patient with right hemisphere language dominance and logopenic-variant primary progressive aphasia (PPA). METHODS Phosphorylated TDP-43 inclusions, neurons, and activated microglia were visualized with immunohistochemical and histologic methods. Markers were quantified bilaterally with unbiased stereology in language- and memory-related cortical regions. RESULTS Clinical MRI indicated cortical atrophy in the right hemisphere, mostly in the temporal lobe. Significantly higher densities of TDP-43 inclusions were present in right language-related temporal regions compared to the left or to other right hemisphere regions. The memory-related entorhinal cortex (ERC) and language regions without significant atrophy showed no asymmetry. Activated microglia displayed extensive asymmetry (R > L). A substantial density of neurons remained in all areas and showed no hemispheric asymmetry. However, perikaryal size was significantly smaller in the right hemisphere across all regions except the ERC. To demonstrate the specificity of this finding, sizes of residual neurons were measured in a right-handed case with PPA and were found to be smaller in the language-dominant left hemisphere. CONCLUSIONS The distribution of TDP-43 inclusions and microglial activation in right temporal language regions showed concordance with anatomic distribution of cortical atrophy and clinical presentation. The results revealed no direct relationship between density of TDP-43 inclusions and activated microglia. Reduced size of the remaining neurons is likely to contribute to cortical atrophy detected by MRI. These findings support the conclusion that there is no obligatory relationship between logopenic PPA and Alzheimer pathology.
Collapse
Affiliation(s)
- Garam Kim
- From the Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Shahrooz Vahedi
- From the Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Tamar Gefen
- From the Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Sandra Weintraub
- From the Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Eileen H Bigio
- From the Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Marek-Marsel Mesulam
- From the Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Changiz Geula
- From the Cognitive Neurology and Alzheimer's Disease Center, Feinberg School of Medicine, Northwestern University, Chicago, IL.
| |
Collapse
|
1626
|
Abstract
Microglia are the resident macrophages of the central nervous system parenchyma and fulfill crucial roles in brain development, homeostasis, and inflammation. The isolation of a pure microglia population from brain tissue enables the examination of microglial phenotypes without the interference of other cell populations. Microglial extractions from the neonatal brain have been described in various protocols, yet the more established and complex adult mouse brain poses a greater challenge. Here we describe a refined protocol including enzymatic and mechanical dissociation of adult mouse brain tissue and removal of myelin by Percoll density gradient. Microglial cells were subsequently extracted by an immunomagnetic approach. This isolation procedure enables the use of functionally viable cells for various applications such as cell culture, flow cytometry, functional assays including bacteria- or bead-based phagocytosis, stimulation assays, and transcriptome profiling techniques such as qRT-PCR and microarray/RNA sequencing.
Collapse
Affiliation(s)
- Kathleen Grabert
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, UK.
| | - Barry W McColl
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, UK
- Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
1627
|
Notter T, Coughlin JM, Sawa A, Meyer U. Reconceptualization of translocator protein as a biomarker of neuroinflammation in psychiatry. Mol Psychiatry 2018; 23:36-47. [PMID: 29203847 DOI: 10.1038/mp.2017.232] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/05/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023]
Abstract
A great deal of interest in psychiatric research is currently centered upon the pathogenic role of inflammatory processes. Positron emission tomography (PET) using radiolabeled ligands selective for the 18 kDa translocator protein (TSPO) has become the most widely used technique to assess putative neuroimmune abnormalities in vivo. Originally used to detect discrete neurotoxic damages, TSPO has generally turned into a biomarker of 'neuroinflammation' or 'microglial activation'. Psychiatric research has mostly accepted these denotations of TSPO, even if they may be inadequate and misleading under many pathological conditions. A reliable and neurobiologically meaningful diagnosis of 'neuroinflammation' or 'microglial activation' is unlikely to be achieved by the sole use of TSPO PET imaging. It is also very likely that the pathological meanings of altered TSPO binding or expression are disease-specific, and therefore, not easily generalizable across different neuropathologies or inflammatory conditions. This difficulty is intricately linked to the varying (and still ill-defined) physiological functions and cellular expression patterns of TSPO in health and disease. While altered TSPO binding or expression may indeed mirror ongoing neuroinflammatory processes in some cases, it may reflect other pathophysiological processes such as abnormalities in cell metabolism, energy production and oxidative stress in others. Hence, the increasing popularity of TSPO PET imaging has paradoxically introduced substantial uncertainty regarding the nature and meaning of neuroinflammatory processes and microglial activation in psychiatry, and likely in other neuropathological conditions as well. The ambiguity of conceiving TSPO simply as a biomarker of 'neuroinflammation' or 'microglial activation' calls for alternative interpretations and complimentary approaches. Without the latter, the ongoing scientific efforts and excitement surrounding the role of the neuroimmune system in psychiatry may not turn into therapeutic hope for affected individuals.
Collapse
Affiliation(s)
- T Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - J M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - U Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
1628
|
Mehrzad J, Hosseinkhani S, Malvandi AM. Human Microglial Cells Undergo Proapoptotic Induction and Inflammatory Activation upon in vitro Exposure to a Naturally Occurring Level of Aflatoxin B1. Neuroimmunomodulation 2018; 25:176-183. [PMID: 30336475 DOI: 10.1159/000493528] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 09/05/2018] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Knowledge regarding interactions of AFB1 with the human nervous system and how a naturally occurring level of AFB1 could potentially induce neuroimmune dysregulation is very limited. To assess the cellular effects of AFB1 on the human brain, we used the human microglia cell line CHME5 as a model to pinpoint its potential in vivo translation. METHODS We used the CHME5 cell line culture system, multiplex qPCR, (chemi)bioluminescence, Luminex ELISA, and flow cytometry assays to evaluate the toxic effects of a naturally occurring level of AFB1 on human microglia. RESULTS A low concentration of AFB1 upregulates the mRNA expression of many proinflammatory molecules, such as TLRs, MyD88, NFκB, and CxCr4, induces intracellular ATP depletion, and increases caspase-3/7 activity at different time points following exposure to the toxin. Furthermore, AFB1-exposed microglia secreted significantly higher levels of IFN-γ and GM-CSF after treatment. We also observed a slight increase in the percentage of apoptotic microglia (annexin V+/PI-) at 48 h posttreatment. CONCLUSION Our work confirmed that the environmentally relevant level of AFB1 could cause an inflammatory reaction in human microglial cells that is potentially harmful or toxic to the homeostasis of the human central nervous system and might increase susceptibility to neurodegenerative diseases.
Collapse
Affiliation(s)
- Jalil Mehrzad
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran,
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Mohammad Malvandi
- International Center for T1D, Pediatric Clinical Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| |
Collapse
|
1629
|
Thion MS, Low D, Silvin A, Chen J, Grisel P, Schulte-Schrepping J, Blecher R, Ulas T, Squarzoni P, Hoeffel G, Coulpier F, Siopi E, David FS, Scholz C, Shihui F, Lum J, Amoyo AA, Larbi A, Poidinger M, Buttgereit A, Lledo PM, Greter M, Chan JKY, Amit I, Beyer M, Schultze JL, Schlitzer A, Pettersson S, Ginhoux F, Garel S. Microbiome Influences Prenatal and Adult Microglia in a Sex-Specific Manner. Cell 2017; 172:500-516.e16. [PMID: 29275859 PMCID: PMC5786503 DOI: 10.1016/j.cell.2017.11.042] [Citation(s) in RCA: 526] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 11/15/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023]
Abstract
Microglia are embryonically seeded macrophages that contribute to brain development, homeostasis, and pathologies. It is thus essential to decipher how microglial properties are temporally regulated by intrinsic and extrinsic factors, such as sexual identity and the microbiome. Here, we found that microglia undergo differentiation phases, discernable by transcriptomic signatures and chromatin accessibility landscapes, which can diverge in adult males and females. Remarkably, the absence of microbiome in germ-free mice had a time and sexually dimorphic impact both prenatally and postnatally: microglia were more profoundly perturbed in male embryos and female adults. Antibiotic treatment of adult mice triggered sexually biased microglial responses revealing both acute and long-term effects of microbiota depletion. Finally, human fetal microglia exhibited significant overlap with the murine transcriptomic signature. Our study shows that microglia respond to environmental challenges in a sex- and time-dependent manner from prenatal stages, with major implications for our understanding of microglial contributions to health and disease. Microglia undergo sequential phases of differentiation during development The maternal microbiome influences microglial properties during prenatal stages The absence of the microbiome has a sex- and time-specific impact on microglia Microbiome depletions have acute and long-term effects on microglial properties
Collapse
Affiliation(s)
- Morgane Sonia Thion
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Aymeric Silvin
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Pauline Grisel
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Jonas Schulte-Schrepping
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Ronnie Blecher
- Department of Immunology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Thomas Ulas
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Paola Squarzoni
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Guillaume Hoeffel
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Aix-Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), 13288 Marseille, France
| | - Fanny Coulpier
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Eleni Siopi
- Institut Pasteur, Unité Perception et Mémoire, CNRS, UMR 3571, F-75015 Paris, France
| | - Friederike Sophie David
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Claus Scholz
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Foo Shihui
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Josephine Lum
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | | | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Anne Buttgereit
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Pierre-Marie Lledo
- Institut Pasteur, Unité Perception et Mémoire, CNRS, UMR 3571, F-75015 Paris, France
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore; KK Research Centre, KK Women's and Children's Hospital, 100 Bukit Timah Road, Singapore 229899, Singapore
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Marc Beyer
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany; Molecular Immunology in Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Joachim Ludwig Schultze
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany; Platform of Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, 53175 Bonn, Germany
| | - Andreas Schlitzer
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore; Myeloid Cell Biology, LIMES-Institute, University of Bonn, 53115 Bonn, Germany
| | - Sven Pettersson
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore; Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17165, Sweden
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore.
| | - Sonia Garel
- Institut de Biologie de l'Ecole normale supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France.
| |
Collapse
|
1630
|
The Role of Microglia and Macrophages in CNS Homeostasis, Autoimmunity, and Cancer. J Immunol Res 2017; 2017:5150678. [PMID: 29410971 PMCID: PMC5749282 DOI: 10.1155/2017/5150678] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 10/20/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022] Open
Abstract
Macrophages are major cell types of the immune system, and they comprise both tissue-resident populations and circulating monocyte-derived subsets. Here, we discuss microglia, the resident macrophage within the central nervous system (CNS), and CNS-infiltrating macrophages. Under steady state, microglia play important roles in the regulation of CNS homeostasis through the removal of damaged or unnecessary neurons and synapses. In the face of inflammatory or pathological insults, microglia and CNS-infiltrating macrophages not only constitute the first line of defense against pathogens by regulating components of innate immunity, but they also regulate the adaptive arms of immune responses. Dysregulation of these responses contributes to many CNS disorders. In this overview, we summarize the current knowledge regarding the highly diverse and complex function of microglia and macrophages during CNS autoimmunity—multiple sclerosis and cancer—malignant glioma. We emphasize how the crosstalk between natural killer (NK) cells or glioma cells or glioma stem cells and CNS macrophages impacts on the pathological processes. Given the essential role of CNS microglia and macrophages in the regulation of all types of CNS disorders, agents targeting these subsets are currently applied in preclinical and clinical trials. We believe that a better understanding of the biology of these macrophage subsets offers new exciting paths for therapeutic intervention.
Collapse
|
1631
|
Chen Y, Yu S, Wu Q, Tang Y, Jiang C, Zhuang Z, Zhao N, Huang B, Xie L. [Effect of bone marrow mesenchymal stem cells conditioned medium on microglia and its secretion of arginase 1 in rats]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2017; 31:1500-1505. [PMID: 29806395 PMCID: PMC8498264 DOI: 10.7507/1002-1892.201710038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 10/30/2017] [Indexed: 11/03/2022]
Abstract
Objective To observe the effect of bone marrow mesenchymal stem cells (BMSCs) conditioned medium on microglia (MGs) and its secretion of arginase 1 (Arg1). Methods The BMSCs separated through differential adhesion method from the femur and tibia marrow of 4-week-old Sprague Dawley (SD) rats were cultured and identified by Vimentin immunofluorescence staining; whereas MGs separated through trypsin digestion method from the brain of 3-day-old SD rats were cultured and identified by Iba1 immunofluorescence staining. The primary MGs were cultured with DMEM/F12 medium containing BMSCs conditioned medium (experimental group) and with single DMEM/F12 medium (control group), respectively. After 48 hours of culture, the morphology of MGs was observed by inverted phase contrast microscope, the activated state of MGs was detected by using Iba1 immunofluorescence staining, and Arg1 expression of MGs was assessed by Iba1-Arg1 double-labelling immunofluorescence staining and Western blot method. Results Inverted phase contrast microscope observation showed that BMSCs entered logarithmic growth phase at 14 days after culture, and more than 98% cells were positive to Vimentin immunofluorescence staining; whereas MGs entered logarithmic growth phase at 21 days after culture, and around 80% cells were positive to Iba1 immunofluorescence staining. Inverted phase contrast microscope observation displayed that in the experimental group, MGs were activated with increased size of soma, shortened process, and amoeba change. Immunofluorescence staining displayed that the Iba1 positive cells number in the experimental group was significantly higher than that in the control group ( t=0.007, P=0.000); double-labelling immunofluorescence staining revealed that the Iba1-Arg1 positive cells number in the experimental group was significantly higher than that in the control group ( t=0.007, P=0.000); and Western blot results elucidated that the relative expression of Arg1 protein in the experimental group was significantly higher than that in the control group ( t=0.001, P=0.000). Conclusion BMSCs conditioned medium can activate MGs and induce MGs to express Arg1.
Collapse
Affiliation(s)
- Yunhui Chen
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Shuguang Yu
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Qiaofeng Wu
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Yong Tang
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Cen Jiang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Zhiqi Zhuang
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Na Zhao
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Biao Huang
- College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China
| | - Lushuang Xie
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075, P.R.China;College of Acupuncture and Moxibustion of Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 610075,
| |
Collapse
|
1632
|
Satoh JI. Gene expression profiles of M1 and M2 microglia characterized by comparative analysis of public datasets. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/cen3.12426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jun-ichi Satoh
- Department of Bioinformatics and Molecular Neuropathology; Meiji Pharmaceutical University; Tokyo Japan
| |
Collapse
|
1633
|
Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer's disease. J Cell Biol 2017; 217:459-472. [PMID: 29196460 PMCID: PMC5800817 DOI: 10.1083/jcb.201709069] [Citation(s) in RCA: 1255] [Impact Index Per Article: 156.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/05/2017] [Accepted: 11/14/2017] [Indexed: 12/15/2022] Open
Abstract
Hansen et al. review the potential dual helpful and harmful roles of microglia in the development and progression of Alzheimer’s disease. Proliferation and activation of microglia in the brain, concentrated around amyloid plaques, is a prominent feature of Alzheimer’s disease (AD). Human genetics data point to a key role for microglia in the pathogenesis of AD. The majority of risk genes for AD are highly expressed (and many are selectively expressed) by microglia in the brain. There is mounting evidence that microglia protect against the incidence of AD, as impaired microglial activities and altered microglial responses to β-amyloid are associated with increased AD risk. On the other hand, there is also abundant evidence that activated microglia can be harmful to neurons. Microglia can mediate synapse loss by engulfment of synapses, likely via a complement-dependent mechanism; they can also exacerbate tau pathology and secrete inflammatory factors that can injure neurons directly or via activation of neurotoxic astrocytes. Gene expression profiles indicate multiple states of microglial activation in neurodegenerative disease settings, which might explain the disparate roles of microglia in the development and progression of AD pathology.
Collapse
Affiliation(s)
- David V Hansen
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA
| | - Jesse E Hanson
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA
| | - Morgan Sheng
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA
| |
Collapse
|
1634
|
Capotondo A, Milazzo R, Garcia-Manteiga JM, Cavalca E, Montepeloso A, Garrison BS, Peviani M, Rossi DJ, Biffi A. Intracerebroventricular delivery of hematopoietic progenitors results in rapid and robust engraftment of microglia-like cells. SCIENCE ADVANCES 2017; 3:e1701211. [PMID: 29226242 PMCID: PMC5721728 DOI: 10.1126/sciadv.1701211] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 11/08/2017] [Indexed: 05/02/2023]
Abstract
Recent evidence indicates that hematopoietic stem and progenitor cells (HSPCs) can serve as vehicles for therapeutic molecular delivery to the brain by contributing to the turnover of resident myeloid cell populations. However, such engraftment needs to be fast and efficient to exert its therapeutic potential for diseases affecting the central nervous system. Moreover, the nature of the cells reconstituted after transplantation and whether they could comprise bona fide microglia remain to be assessed. We demonstrate that transplantation of HSPCs in the cerebral lateral ventricles provides rapid engraftment of morphologically, antigenically, and transcriptionally dependable microglia-like cells. We show that the cells comprised within the hematopoietic stem cell compartment and enriched early progenitor fractions generate this microglia-like population when injected in the brain ventricles in the absence of engraftment in the bone marrow. This delivery route has therapeutic relevance because it increases the delivery of therapeutic molecules to the brain, as shown in a humanized animal model of a prototypical lysosomal storage disease affecting the central nervous system.
Collapse
Affiliation(s)
- Alessia Capotondo
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milano, Italy
| | - Rita Milazzo
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milano, Italy
| | - Jose M. Garcia-Manteiga
- Centre for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Eleonora Cavalca
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milano, Italy
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA 02115, USA
| | - Annita Montepeloso
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milano, Italy
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA 02115, USA
| | - Brian S. Garrison
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Program in Cellular and Molecular Medicine, Department of Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Marco Peviani
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milano, Italy
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA 02115, USA
| | - Derrick J. Rossi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Program in Cellular and Molecular Medicine, Department of Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Alessandra Biffi
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milano, Italy
- Gene Therapy Program, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA 02115, USA
- Gene Therapy Program, Department of Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Corresponding author.
| |
Collapse
|
1635
|
Loss of APOBEC1 RNA-editing function in microglia exacerbates age-related CNS pathophysiology. Proc Natl Acad Sci U S A 2017; 114:13272-13277. [PMID: 29167375 DOI: 10.1073/pnas.1710493114] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Microglia (MG), a heterogeneous population of phagocytic cells, play important roles in central nervous system (CNS) homeostasis and neural plasticity. Under steady-state conditions, MG maintain homeostasis by producing antiinflammatory cytokines and neurotrophic factors, support myelin production, and remove synapses and cellular debris, as well as participating in "cross-correction," a process that supplies neurons with key factors for executing autophagy-lysosomal function. As sentinels for the immune system, MG also detect "danger" signals (pathogenic or traumatic insult), become activated, produce proinflammatory cytokines, and recruit monocytes and dendritic cells to the site of damage through a breached blood-brain barrier or via brain lymphatics. Failure to effectively resolve MG activation can be problematic and can lead to chronic inflammation, a condition proposed to underlie CNS pathophysiology in heritable brain disorders and age-related neurodegenerative and cognitive decline. Here, we show that APOBEC1-mediated RNA editing occurs within MG and is key to maintaining their resting status. Like bone marrow-derived macrophages, RNA editing in MG leads to overall changes in the abundance of edited proteins that coordinate the function of multiple cellular pathways. Conversely, mice lacking the APOBEC1 editing function in MG display evidence of dysregulation, with progressive age-related signs of neurodegeneration, characterized by clustering of activated MG, aberrant myelination, increased inflammation, and lysosomal anomalies that culminate in behavioral and motor deficiencies. Collectively, our study identifies posttranscriptional modification by RNA editing as a critical regulatory mechanism of vital cellular functions that maintain overall brain health.
Collapse
|
1636
|
Abstract
Microglia and non-parenchymal macrophages in the brain are mononuclear phagocytes that are increasingly recognized to be essential players in the development, homeostasis and diseases of the central nervous system. With the availability of new genetic, molecular and pharmacological tools, considerable advances have been made towards our understanding of the embryonic origins, developmental programmes and functions of these cells. These exciting discoveries, some of which are still controversial, also raise many new questions, which makes brain macrophage biology a fast-growing field at the intersection of neuroscience and immunology. Here, we review the current knowledge of how and where brain macrophages are generated, with a focus on parenchymal microglia. We also discuss their normal functions during development and homeostasis, the disturbance of which may lead to various neurodegenerative and neuropsychiatric diseases.
Collapse
|
1637
|
Liu Y, Xie X, Xia LP, Lv H, Lou F, Ren Y, He ZY, Luo XG. Peripheral immune tolerance alleviates the intracranial lipopolysaccharide injection-induced neuroinflammation and protects the dopaminergic neurons from neuroinflammation-related neurotoxicity. J Neuroinflammation 2017; 14:223. [PMID: 29145874 PMCID: PMC5693474 DOI: 10.1186/s12974-017-0994-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neuroinflammation plays a critical role in the onset and development of neurodegeneration disorders such as Parkinson's disease. The immune activities of the central nervous system are profoundly affected by peripheral immune activities. Immune tolerance refers to the unresponsiveness of the immune system to continuous or repeated stimulation to avoid excessive inflammation and unnecessary by-stander injury in the face of continuous antigen threat. It has been proved that the immune tolerance could suppress the development of various peripheral inflammation-related diseases. However, the role of immune tolerance in neuroinflammation and neurodegenerative diseases was not clear. METHODS Rats were injected with repeated low-dose lipopolysaccharide (LPS, 0.3 mg/kg) intraperitoneally for 4 days to induce peripheral immune tolerance. Neuroinflammation was produced using intracranial LPS (15 μg) injection. Inflammation cytokines were measured using enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). Microglial activation were measured using immunostaining of Iba-1 and ED-1. Dopaminergic neuronal damage was evaluated using immunochemistry staining and stereological counting of TH-positive neurons. Behavioral impairment was evaluated using amphetamine-induced rotational behavioral assessment. RESULTS Compared with the non-immune tolerated animals, pre-treatment of peripheral immune tolerance significantly decreased the production of inflammatory cytokines, suppressed the microglial activation, and increased the number of dopaminergic neuronal survival in the substantia nigra. CONCLUSIONS Our results indicated that peripheral immune tolerance attenuated neuroinflammation and inhibited neuroinflammation-induced dopaminergic neuronal death.
Collapse
Affiliation(s)
- Yang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Xin Xie
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Li-Ping Xia
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Hong Lv
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Fan Lou
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Yan Ren
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Zhi-Yi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Xiao-Guang Luo
- Department of Neurology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
1638
|
Alawieyah Syed Mortadza S, Sim JA, Neubrand VE, Jiang LH. A critical role of TRPM2 channel in Aβ42
-induced microglial activation and generation of tumor necrosis factor-α. Glia 2017; 66:562-575. [DOI: 10.1002/glia.23265] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/26/2017] [Accepted: 11/03/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Sharifah Alawieyah Syed Mortadza
- School of Biomedical Sciences, Faculty of Biological Sciences; University of Leeds; Leeds UK
- Faculty of Medicine and Health Science; University Putra Malaysia; Selangor Malaysia
| | - Joan A. Sim
- School of Medicine; University of Manchester; Manchester UK
| | - Veronika E. Neubrand
- Department of Cell Biology and Immunology, IPBLN-CSIC, Avda Conocimiento, PT Ciencias de La Salud; Institute of Parasitology and Biomedicine López-Neyra; Granada Spain
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences; University of Leeds; Leeds UK
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province and Department of Physiology and Neurobiology; Xinxiang Medical University; Xinxiang China
| |
Collapse
|
1639
|
Nadal-Nicolás FM, Jiménez-López M, Salinas-Navarro M, Sobrado-Calvo P, Vidal-Sanz M, Agudo-Barriuso M. Microglial dynamics after axotomy-induced retinal ganglion cell death. J Neuroinflammation 2017; 14:218. [PMID: 29121969 PMCID: PMC5679427 DOI: 10.1186/s12974-017-0982-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/16/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Microglial cells (MCs) are the sentries of the central nervous system. In health, they are known as surveying MCs because they examine the tissue to maintain the homeostasis. In disease, they activate and, among other functions, become phagocytic to clean the cellular debris. In this work, we have studied the behavior of rat retinal MCs in two models of unilateral complete intraorbital optic nerve axotomy which elicit a different time course of retinal ganglion cell (RGC) loss. METHODS Albino Sprague-Dawley rats were divided into these groups: (a) intact (no surgery), (b) fluorogold (FG) tracing from the superior colliculi, and (c) FG tracing + crush or transection of the left optic nerve. The retinas were dissected from 2 days to 2 months after the lesions (n = 4-12 group/lesion and time point) and then were subjected to Brn3a and Iba1 double immunodetection. In each intact retina, the total number of Brn3a+RGCs and Iba+MCs was quantified. In each traced retina (b and c groups), FG-traced RGCs and phagocytic microglial cells (PMCs, FG+Iba+) were also quantified. Topographical distribution was assessed by neighbor maps. RESULTS In intact retinas, surveying MCs are homogenously distributed in the ganglion cell layer and the inner plexiform layer. Independently of the axotomy model, RGC death occurs in two phases, one quick and one protracted, and there is a lineal and topographical correlation between the appearance of PMCs and the loss of traced RGCs. Furthermore, the clearance of FG+RGCs by PMCs occurs 3 days after the actual loss of Brn3a expression that marks RGC death. In addition, almost 50% of MCs from the inner plexiform layer migrate to the ganglion cell layer during the quick phase of RGC loss, returning to the inner plexiform layer during the slow degeneration phase. Finally, in contrast to what happens in mice, in rats, there is no microglial phagocytosis in the contralateral uninjured retina. CONCLUSIONS Axotomy-induced RGC death occurs earlier than RGC clearance and there is an inverse correlation between RGC loss and PMC appearance, both numerically and topographically, suggesting that phagocytosis occurs as a direct response to RGC death rather than to axonal damage.
Collapse
Affiliation(s)
- Francisco M Nadal-Nicolás
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca, Edificio LAIB Planta 5ª, Carretera Buenavista s/n, 30120, El Palmar, Murcia, Spain.
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain.
- Present address: Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Manuel Jiménez-López
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca, Edificio LAIB Planta 5ª, Carretera Buenavista s/n, 30120, El Palmar, Murcia, Spain
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - Manuel Salinas-Navarro
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca, Edificio LAIB Planta 5ª, Carretera Buenavista s/n, 30120, El Palmar, Murcia, Spain
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - Paloma Sobrado-Calvo
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca, Edificio LAIB Planta 5ª, Carretera Buenavista s/n, 30120, El Palmar, Murcia, Spain
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - Manuel Vidal-Sanz
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca, Edificio LAIB Planta 5ª, Carretera Buenavista s/n, 30120, El Palmar, Murcia, Spain
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - Marta Agudo-Barriuso
- Grupo de Oftalmología Experimental, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca, Edificio LAIB Planta 5ª, Carretera Buenavista s/n, 30120, El Palmar, Murcia, Spain.
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain.
| |
Collapse
|
1640
|
Abstract
The innate immune system plays diverse roles in health and disease. It represents the first line of defense against infection and is involved in tissue repair, wound healing, and clearance of apoptotic cells and cellular debris. Excessive or nonresolving innate immune activation can lead to systemic or local inflammatory complications and cause or contribute to the development of inflammatory diseases. In the brain, microglia represent the key innate immune cells, which are involved in brain development, brain maturation, and homeostasis. Impaired microglial function, either through aberrant activation or decreased functionality, can occur during aging and during neurodegeneration, and the resulting inflammation is thought to contribute to neurodegenerative diseases. This review highlights recent advances in our understanding of the influence of innate immunity on neurodegenerative disorders such as Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease, and Huntington's disease.
Collapse
Affiliation(s)
- Larisa I Labzin
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom;
| | - Michael T Heneka
- Department of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University Hospitals Bonn, Bonn 53127, Germany; .,Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.,German Center for Neurodegenerative Diseases, Bonn 53175, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospitals Bonn, Bonn 53127, Germany; .,Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.,German Center for Neurodegenerative Diseases, Bonn 53175, Germany.,Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim 7491, Norway
| |
Collapse
|
1641
|
Satoh JI, Kino Y, Yanaizu M, Tosaki Y, Sakai K, Ishida T, Saito Y. Microglia express ABI3 in the brains of Alzheimer's disease and Nasu-Hakola disease. Intractable Rare Dis Res 2017; 6:262-268. [PMID: 29259854 PMCID: PMC5735279 DOI: 10.5582/irdr.2017.01073] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Nasu-Hakola disease (NHD) is a rare autosomal recessive leukoencephalopathy caused by a loss-of-function mutation of either TYROBP (DAP12) or TREM2 expressed in microglia. A rare variant of the TREM2 gene encoding p.Arg47His causes a 3-fold increase in the risk for late-onset Alzheimer's disease (LOAD). A recent study demonstrated that a rare coding variant p.Ser209Phe in the ABI family member 3 (ABI3) gene, a regulator of actin cytoskeleton organization, confers risk of developing of LOAD, although the pattern of ABI3 expression in AD and NHD brains with relevance to microglial pathology remains to be characterized. We investigated the cell type-specific expression of ABI3 in the brains derived from four non-neurological controls (NC), ten AD and five NHD cases by immunohistochemistry. We identified an intense ABI3 immunoreactivity chiefly on a subset of microglia with ramified or amoeboid morphology located in the grey matter and the white matter of the frontal cortex and the hippocampus of NC, AD, and NHD cases. The immunolabeled area of ABI3-positive microglia was not significantly different among NC, AD, and NHD cases due to great variability from case to case. The clusters of ABI3-immunoreactive microglia were found exclusively in AD brains and they were associated with amyloid plaques. Although these observations do not actively support the view that ABI3-immunoreactive microglia play a central role in the development of leukoencephalopathy in NHD brains and the neurodegeneration in AD brains, the intense expression of ABI3 on microglia might regulate their migration under conditions of health and disease in the central nervous system (CNS).
Collapse
Affiliation(s)
- Jun-ichi Satoh
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Tokyo, Japan
- Address correspondence to: Dr. Jun-ichi Satoh, Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, Japan. E-mail:
| | - Yoshihiro Kino
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Motoaki Yanaizu
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Youhei Tosaki
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Kenji Sakai
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Tsuyoshi Ishida
- Department of Pathology and Laboratory Medicine, Kohnodai Hospital, NCGM, Chiba, Japan
| | - Yuko Saito
- Department of Laboratory Medicine, National Center Hospital, NCNP, Tokyo, Japan
| |
Collapse
|
1642
|
Walter J, Kemmerling N, Wunderlich P, Glebov K. γ-Secretase in microglia - implications for neurodegeneration and neuroinflammation. J Neurochem 2017; 143:445-454. [PMID: 28940294 DOI: 10.1111/jnc.14224] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/16/2017] [Accepted: 09/18/2017] [Indexed: 12/13/2022]
Abstract
γ-Secretase is an intramembrane cleaving protease involved in the generation of the Alzheimer's disease (AD)-associated amyloid β peptide (Aβ). γ-Secretase is ubiquitously expressed in different organs, and also in different cell types of the human brain. Besides the involvement in the proteolytic generation of Aβ from the amyloid precursor protein, γ-secretase cleaves many additional protein substrates, suggesting pleiotropic functions under physiological and pathophysiological conditions. Microglia exert important functions during brain development and homeostasis in adulthood, and accumulating evidence indicates that microglia and neuroinflammatory processes contribute to the pathogenesis of neurodegenerative diseases. Recent studies demonstrate functional implications of γ-secretase in microglia, suggesting that alterations in γ-secretase activity could contribute to AD pathogenesis by modulation of microglia and related neuroinflammatory processes during neurodegeneration. In this review, we discuss the involvement of γ-secretase in the regulation of microglial functions, and the potential relevance of these processes under physiological and pathophysiological conditions. This article is part of the series "Beyond Amyloid".
Collapse
Affiliation(s)
- Jochen Walter
- Department of Neurology, University of Bonn, Bonn, Germany
| | | | | | | |
Collapse
|
1643
|
Zhou T, Huang Z, Sun X, Zhu X, Zhou L, Li M, Cheng B, Liu X, He C. Microglia Polarization with M1/M2 Phenotype Changes in rd1 Mouse Model of Retinal Degeneration. Front Neuroanat 2017; 11:77. [PMID: 28928639 PMCID: PMC5591873 DOI: 10.3389/fnana.2017.00077] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 08/21/2017] [Indexed: 01/28/2023] Open
Abstract
Microglia activation is recognized as the hallmark of neuroinflammation. However, the activation profile and phenotype changes of microglia during the process of retinal degeneration are poorly understood. This study aimed to elucidate the time-spatial pattern of microglia distribution and characterize the polarized phenotype of activated microglia during retinal neuroinflammation and degeneration in rd1 (Pde6βrd1/rd1) mice, the classic model of inherited retinal degeneration. Retinae of rd1 mice at different postnatal days (P7, P14, P21, P28, P56, and P180) were prepared for further analysis. We found most CD11b+ or IBA1+ microglia expressed Ki-67 and CD68 in rd1 mice and these cells migrated toward the layer of degenerative photoreceptors at the rapid rods degeneration phase from P14 to P28. These microglia exhibited typical ameboid activated shape with round bodies and scarce dendrites, while at late phase at P180, they displayed resting ramified morphology with elongated dendrites. Flow cytometry revealed that the percentage of CD86+CD206- M1 microglia increased markedly in rd1 retinae, however, no significant change was observed in CD206+CD86- M2 microglia. Interestingly, CD86+CD206+ microglia, an intermediate state between the two extremes of M1 and M2, increased markedly at the rapid rods degeneration phase. The immunofluorescence images revealed that microglia in rd1 mice highly expressed M1 markers including CD16/32, CD86, and CD40. In addition, increased expression of pro-inflammatory cytokines (TNF-α, IL-6, and CCL2) was observed in rd1 mice. Our findings unfolded a panorama for the first time that microglia conducted distinctive behaviors with the progression of retinal degeneration in rd1 mice. Microglia is activated and particularly polarized to a pro-inflammatory M1 phenotype at the rapid rods degenerative phase, suggesting that the involvement of M1 microglia in the retinal neuroinflammation and degeneration. Most microglia adopted an intermediate polarization “M1½” state in rd1, revealing that microglia orchestrated a complicated continuous spectrum in degenerative retina.
Collapse
Affiliation(s)
- Tian Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Zijing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Xiaowei Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Xiaowei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Lingli Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Mei Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Bing Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| | - Chang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
1644
|
Microglia contribute to normal myelinogenesis and to oligodendrocyte progenitor maintenance during adulthood. Acta Neuropathol 2017; 134:441-458. [PMID: 28685323 DOI: 10.1007/s00401-017-1747-1] [Citation(s) in RCA: 374] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/14/2017] [Accepted: 07/01/2017] [Indexed: 12/11/2022]
Abstract
Whereas microglia involvement in virtually all brain diseases is well accepted their role in the control of homeostasis in the central nervous system (CNS) is mainly thought to be the maintenance of neuronal function through the formation, refinement, and monitoring of synapses in both the developing and adult brain. Although the prenatal origin as well as the neuron-centered function of cortical microglia has recently been elucidated, much less is known about a distinct amoeboid microglia population formerly described as the "fountain of microglia" that appears only postnatally in myelinated regions such as corpus callosum and cerebellum. Using large-scale transcriptional profiling, fate mapping, and genetic targeting approaches, we identified a unique molecular signature of this microglia subset that arose from a CNS endogenous microglia pool independent from circulating myeloid cells. Microglia depletion experiments revealed an essential role of postnatal microglia for the proper development and homeostasis of oligodendrocytes and their progenitors. Our data provide new cellular and molecular insights into the myelin-supporting function of microglia in the normal CNS.
Collapse
|
1645
|
Abstract
Microglia are brain-resident myeloid cells that mediate key functions to support the CNS. Microglia express a wide range of receptors that act as molecular sensors, which recognize exogenous or endogenous CNS insults and initiate an immune response. In addition to their classical immune cell function, microglia act as guardians of the brain by promoting phagocytic clearance and providing trophic support to ensure tissue repair and maintain cerebral homeostasis. Conditions associated with loss of homeostasis or tissue changes induce several dynamic microglial processes, including changes of cellular morphology, surface phenotype, secretory mediators, and proliferative responses (referred to as an "activated state"). Activated microglia represent a common pathological feature of several neurodegenerative diseases, including Alzheimer's disease (AD). Cumulative evidence suggests that microglial inflammatory activity in AD is increased while microglial-mediated clearance mechanisms are compromised. Microglia are perpetually engaged in a mutual interaction with the surrounding environment in CNS; thus, diverse microglial reactions at different disease stages may open new avenues for therapeutic intervention and modification of inflammatory activities. In this Review, the role of microglia in the pathogenesis of AD and the modulation of microglia activity as a therapeutic modality will be discussed.
Collapse
Affiliation(s)
- Heela Sarlus
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany.,Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
1646
|
Are Astrocytes the Predominant Cell Type for Activation of Nrf2 in Aging and Neurodegeneration? Antioxidants (Basel) 2017; 6:antiox6030065. [PMID: 28820437 PMCID: PMC5618093 DOI: 10.3390/antiox6030065] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 12/29/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that regulates hundreds of antioxidant genes, and is activated in response to oxidative stress. Given that many neurodegenerative diseases including Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, Huntington’s disease and multiple sclerosis are characterised by oxidative stress, Nrf2 is commonly activated in these diseases. Evidence demonstrates that Nrf2 activity is repressed in neurons in vitro, and only cultured astrocytes respond strongly to Nrf2 inducers, leading to the interpretation that Nrf2 signalling is largely restricted to astrocytes. However, Nrf2 activity can be observed in neurons in post-mortem brain tissue and animal models of disease. Thus this interpretation may be false, and a detailed analysis of the cell type expression of Nrf2 in neurodegenerative diseases is required. This review describes the evidence for Nrf2 activation in each cell type in prominent neurodegenerative diseases and normal aging in human brain and animal models of neurodegeneration, the response to pharmacological and genetic modulation of Nrf2, and clinical trials involving Nrf2-modifying drugs.
Collapse
|
1647
|
Tikamdas R, Singhal S, Zhang P, Smith JA, Krause EG, Stevens SM, Song S, Liu B. Ischemia-responsive protein 94 is a key mediator of ischemic neuronal injury-induced microglial activation. J Neurochem 2017. [PMID: 28640931 DOI: 10.1111/jnc.14111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Neuroinflammation, especially activation of microglia, the key immune cells in the brain, has been proposed to contribute to the pathogenesis of ischemic stroke. However, the dynamics and the potential mediators of microglial activation following ischemic neuronal injury are not well understood. In this study, using oxygen/glucose deprivation and reoxygenation with neuronal and microglial cell cultures as an in vitro model of ischemic neuronal injury, we set out to identify neuronal factors released from injured neurons that are capable of inducing microglial activation. Conditioned media (CM) from hippocampal and cortical neurons exposed to oxygen/glucose deprivation and reoxygenation induced significant activation of microglial cells as well as primary microglia, evidenced by up-regulation of inducible nitric oxide synthase, increased production of nitrite and reactive oxygen species, and increased expression of microglial markers. Mechanistically, neuronal ischemia-responsive protein 94 (Irp94) was a key contributor to microglial activation since significant increase in Irp94 was detected in the neuronal CM following ischemic insult and immunodepletion of Irp94 rendered ischemic neuronal CM ineffective in inducing microglial activation. Ischemic insult-augmented oxidative stress was a major facilitator of neuronal Irp94 release, and pharmacological inhibition of NADPH oxidase significantly reduced the ischemic injury-induced neuronal reactive oxygen species production and Irp94 release. Taken together, these results indicate that neuronal Irp94 may play a pivotal role in the propagation of ischemic neuronal damage. Continued studies may help identify Irp94 and/or related proteins as potential therapeutic targets and/or diagnostic/prognostic biomarkers for managing ischemia-associated brain disorders.
Collapse
Affiliation(s)
- Rajiv Tikamdas
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Sarthak Singhal
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Ping Zhang
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Justin A Smith
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Eric G Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Stanley M Stevens
- Department of Cell Biology, Microbiology and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, Florida, USA
| | - Sihong Song
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Bin Liu
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
1648
|
Klein RS, Hunter CA. Protective and Pathological Immunity during Central Nervous System Infections. Immunity 2017; 46:891-909. [PMID: 28636958 PMCID: PMC5662000 DOI: 10.1016/j.immuni.2017.06.012] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/05/2017] [Accepted: 06/05/2017] [Indexed: 02/08/2023]
Abstract
The concept of immune privilege of the central nervous system (CNS) has dominated the study of inflammatory processes in the brain. However, clinically relevant models have highlighted that innate pathways limit pathogen invasion of the CNS and adaptive immunity mediates control of many neural infections. As protective responses can result in bystander damage, there are regulatory mechanisms that balance protective and pathological inflammation, but these mechanisms might also allow microbial persistence. The focus of this review is to consider the host-pathogen interactions that influence neurotropic infections and to highlight advances in our understanding of innate and adaptive mechanisms of resistance as key determinants of the outcome of CNS infection. Advances in these areas have broadened our comprehension of how the immune system functions in the brain and can readily overcome immune privilege.
Collapse
Affiliation(s)
- Robyn S Klein
- Departments of Medicine, Pathology and Immunology, Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
1649
|
Abstract
Microglia are the principal resident immune cells in the central nervous system and are believed to be versatile players in both inflammatory and physiological contexts. On the one hand, in order to safeguard the microenvironment microglia can be rapidly activated by contact with microbial products or cell debris, thereby exerting the functions of innate immunity via phagocytosis and secretion of cytokines and chemokines. Conversely, microglia can also assist in brain development, synaptic plasticity and neural repair through the production of neurotrophic factors and clearance of myelin debris. It is now well accepted that the dysfunction of microglia and microglia-induced neuroinflammation are implicated in the occurrence and progression of many neurological diseases. Although the past decade has witnessed major progress in understanding of multi-tasking microglia, what remains largely enigmatic is the relative importance of microglia at different disease stages and how microglia should be targeted for optimal therapeutic efficacy. Notably, microglia depletion through genetic targeting or pharmacological therapies can be viewed as effective tools to stimulate new microglia to repopulate the central nervous system. Microglia depletion and subsequent repopulation at defined stages in various experimental animal model disorders allow us to extend our knowledge of molecular mechanisms, thus holding promise for designing strategies to resolve neuroinflammation and promote recovery. Herein we highlight the highly plastic and diverse phenotypes of microglia and outline the lessons learned from microglia depletion approaches.
Collapse
Affiliation(s)
- Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at Solna, CMM L8:04, Karolinska Sjukhuset, S-171 76, Stockholm, Sweden
| | - Robert A Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at Solna, CMM L8:04, Karolinska Sjukhuset, S-171 76, Stockholm, Sweden
| | - Xing-Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at Solna, CMM L8:04, Karolinska Sjukhuset, S-171 76, Stockholm, Sweden.
| |
Collapse
|
1650
|
Salvi V, Sozio F, Sozzani S, Del Prete A. Role of Atypical Chemokine Receptors in Microglial Activation and Polarization. Front Aging Neurosci 2017; 9:148. [PMID: 28603493 PMCID: PMC5445112 DOI: 10.3389/fnagi.2017.00148] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/02/2017] [Indexed: 01/07/2023] Open
Abstract
Inflammatory reactions occurring in the central nervous system (CNS), known as neuroinflammation, are key components of the pathogenic mechanisms underlying several neurological diseases. The chemokine system plays a crucial role in the recruitment and activation of immune and non-immune cells in the brain, as well as in the regulation of microglia phenotype and function. Chemokines belong to a heterogeneous family of chemotactic agonists that signal through the interaction with G protein-coupled receptors (GPCRs). Recently, a small subset of chemokine receptors, now identified as “atypical chemokine receptors” (ACKRs), has been described. These receptors lack classic GPCR signaling and chemotactic activity and are believed to limit inflammation through their ability to scavenge chemokines at the inflammatory sites. Recent studies have highlighted a role for ACKRs in neuroinflammation. However, in the CNS, the role of ACKRs seems to be more complex than the simple control of inflammation. For instance, CXCR7/ACKR3 was shown to control T cell trafficking through the regulation of CXCL12 internalization at CNS endothelial barriers. Furthermore, D6/ACKR2 KO mice were protected in a model of experimental autoimmune encephalomyelitis (EAE). D6/ACKR2 KO showed an abnormal accumulation of dendritic cells at the immunization and a subsequent impairment in T cell priming. Finally, CCRL2, an ACKR-related protein, was shown to play a role in the control of the resolution phase of EAE. Indeed, CCRL2 KO mice showed exacerbated, non-resolving disease with protracted inflammation and increased demyelination. This phenotype was associated with increased microglia and macrophage activation markers and imbalanced M1 vs. M2 polarization. This review will summarize the current knowledge on the role of the ACKRs in neuroinflammation with a particular attention to their role in microglial polarization and function.
Collapse
Affiliation(s)
- Valentina Salvi
- Department of Molecular and Translational Medicine, University of BresciaBrescia, Italy
| | - Francesca Sozio
- Department of Molecular and Translational Medicine, University of BresciaBrescia, Italy.,IRCCS-Humanitas Clinical and Research CenterRozzano-Milano, Italy
| | - Silvano Sozzani
- Department of Molecular and Translational Medicine, University of BresciaBrescia, Italy.,IRCCS-Humanitas Clinical and Research CenterRozzano-Milano, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of BresciaBrescia, Italy.,IRCCS-Humanitas Clinical and Research CenterRozzano-Milano, Italy
| |
Collapse
|