151
|
ATG-dependent phagocytosis in dendritic cells drives myelin-specific CD4 + T cell pathogenicity during CNS inflammation. Proc Natl Acad Sci U S A 2017; 114:E11228-E11237. [PMID: 29233943 DOI: 10.1073/pnas.1713664114] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Although reactivation and accumulation of autoreactive CD4+ T cells within the CNS are considered to play a key role in the pathogenesis of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), the mechanisms of how these cells recognize their target organ and induce sustained inflammation are incompletely understood. Here, we report that mice with conditional deletion of the essential autophagy protein ATG5 in classical dendritic cells (DCs), which are present at low frequencies in the nondiseased CNS, are completely resistant to EAE development following adoptive transfer of myelin-specific T cells and show substantially reduced in situ CD4+ T cell accumulation during the effector phase of the disease. Endogenous myelin peptide presentation to CD4+ T cells following phagocytosis of injured, phosphatidylserine-exposing oligodendroglial cells is abrogated in the absence of ATG5. Pharmacological inhibition of ATG-dependent phagocytosis by the cardiac glycoside neriifolin, an inhibitor of the Na+, K+-ATPase, delays the onset and reduces the clinical severity of EAE induced by myelin-specific CD4+ T cells. These findings link phagocytosis of injured oligodendrocytes, a pathological hallmark of MS lesions and during EAE, with myelin antigen processing and T cell pathogenicity, and identify ATG-dependent phagocytosis in DCs as a key regulator in driving autoimmune CD4+ T cell-mediated CNS damage.
Collapse
|
152
|
Host STING-dependent MDSC mobilization drives extrinsic radiation resistance. Nat Commun 2017; 8:1736. [PMID: 29170400 PMCID: PMC5701019 DOI: 10.1038/s41467-017-01566-5] [Citation(s) in RCA: 313] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 09/29/2017] [Indexed: 11/08/2022] Open
Abstract
Radiotherapy induces and promotes innate and adaptive immunity in which host STING plays an important role. However, radioresistance in irradiated tumors can also develop, resulting in relapse. Here we report a mechanism by which extrinsic resistance develops after local ablative radiation that relies on the immunosuppressive action of STING. The STING/type I interferon pathway enhances suppressive inflammation in tumors by recruiting myeloid cells in part via the CCR2 pathway. Germ-line knockouts of CCR2 or treatment with an anti-CCR2 antibody results in blockade of radiation-induced MDSC infiltration. Treatment with anti-CCR2 antibody alleviates immunosuppression following activation of the STING pathway, enhancing the anti-tumor effects of STING agonists and radiotherapy. We propose that radiation-induced STING activation is immunosuppressive due to (monocytic) M-MDSC infiltration, which results in tumor radioresistance. Furthermore, the immunosuppressive effects of radiotherapy and STING agonists can be abrogated in humans by a translational strategy involving anti-CCR2 antibody treatment to improve radiotherapy.
Collapse
|
153
|
Cédile O, Wlodarczyk A, Owens T. CCL2 recruits T cells into the brain in a CCR2-independent manner. APMIS 2017; 125:945-956. [PMID: 28836736 DOI: 10.1111/apm.12740] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/09/2017] [Indexed: 12/24/2022]
Abstract
CCL2 is a chemokine that can be induced during neuroinflammation to recruit immune cells, but its role in the central nervous system (CNS) is unclear. Our aim was to better understand its role. We induced CCL2 in CNS of naive CCL2-deficient mice using intrathecally administered replication-defective adenovirus and examined cell infiltration by flow cytometry. CCL2 expression induced pronounced and unexpected recruitment of regulatory and IFNγ-producing T cells to CNS from blood, possibly related to defective egress of monocytes from CCL2-deficient bone marrow. Infiltration also occurred in mice lacking CCR2, a receptor for CCL2. Expression of another receptor for CCL2, CCR4, and CXCR3, a receptor for CXCL10, which was also induced, were both increased in CCL2-treated CNS. CCR4 was expressed by neurons and astrocytes as well as CD4 T cells, and CXCR3 was expressed by CD4 and CD8 T cells. Chemokine-recruited T cells did not lead to CNS pathology. Our findings show a role for CCL2 in recruitment of CD4 T cells to the CNS and show that redundancy among chemokine receptors ensures optimal response.
Collapse
Affiliation(s)
- Oriane Cédile
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
154
|
The role of the immune system in Alzheimer disease: Etiology and treatment. Ageing Res Rev 2017; 40:84-94. [PMID: 28941639 DOI: 10.1016/j.arr.2017.08.005] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/25/2017] [Accepted: 08/31/2017] [Indexed: 12/24/2022]
Abstract
The immune system is now considered a major factor in Alzheimer Disease (AD). This review seeks to demonstrate how various aspects of the immune system, both in the brain and peripherally, interact to contribute to AD. We highlight classical nervous system immune components, such as complement and microglia, as well as novel aspects of the peripheral immune system that can influence disease, such as monocytes and lymphocytes. By detailing the roles of various immune cells in AD, we summarize an emerging perspective for disease etiology and future therapeutic targets.
Collapse
|
155
|
Lloyd AF, Davies CL, Miron VE. Microglia: origins, homeostasis, and roles in myelin repair. Curr Opin Neurobiol 2017; 47:113-120. [PMID: 29073528 DOI: 10.1016/j.conb.2017.10.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/26/2017] [Accepted: 10/07/2017] [Indexed: 11/27/2022]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS), implicated in developmental processes, homeostasis, and responses to injury. Derived from the yolk sac during development, microglia self-renew, self-regulate their numbers during homeostatic conditions, and show a robust proliferative capacity even in adulthood. Together with monocyte-derived macrophages (MDM), microglia coordinate the regeneration of CNS myelin around axons, termed remyelination. Gene expression analyses and experimental modelling have identified pro-remyelination roles for microglia/MDM in clearance of myelin debris, secretion of growth factors, and remodelling of the extracellular matrix. Further investigations into the molecular mechanisms controlling these regenerative functions will reveal novel therapeutic strategies to enhance remyelination, by harnessing the beneficial effects of the innate immune response to injury.
Collapse
Affiliation(s)
- Amy F Lloyd
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | - Claire L Davies
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| |
Collapse
|
156
|
Differential contribution of microglia and monocytes in neurodegenerative diseases. J Neural Transm (Vienna) 2017; 125:809-826. [PMID: 29063348 DOI: 10.1007/s00702-017-1795-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 10/03/2017] [Indexed: 12/12/2022]
Abstract
Neuroinflammation is a hallmark of neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Microglia, the innate immune cells of the CNS, are the first to react to pathological insults. However, multiple studies have also demonstrated an involvement of peripheral monocytes in several neurodegenerative diseases. Due to the different origins of these two cell types, it is important to distinguish their role and function in the development and progression of these diseases. In this review, we will summarize and discuss the current knowledge of the differential contributions of microglia and monocytes in the common neurodegenerative diseases AD, PD, and ALS, as well as multiple sclerosis, which is now regarded as a combination of inflammatory processes and neurodegeneration. Until recently, it has been challenging to differentiate microglia from monocytes, as there were no specific markers. Therefore, the recent identification of specific molecular signatures of both cell types will help to advance our understanding of their differential contribution in neurodegenerative diseases.
Collapse
|
157
|
Montes-Cobos E, Schweingruber N, Li X, Fischer HJ, Reichardt HM, Lühder F. Deletion of the Mineralocorticoid Receptor in Myeloid Cells Attenuates Central Nervous System Autoimmunity. Front Immunol 2017; 8:1319. [PMID: 29081780 PMCID: PMC5645513 DOI: 10.3389/fimmu.2017.01319] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022] Open
Abstract
Myeloid cells play an important role in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Monocytes, macrophages, and microglia can adopt two distinct phenotypes, with M1-polarized cells being more related to inflammation and autoimmunity while M2-polarized cells contribute to tissue repair and anti-inflammatory processes. Here, we show that deletion of the mineralocorticoid receptor (MR) in bone marrow-derived macrophages and peritoneal macrophages caused their polarization toward the M2 phenotype with its distinct gene expression, altered phagocytic and migratory properties, and dampened NO production. After induction of EAE, mice that are selectively devoid of the MR in their myeloid cells (MRlysM mice) showed diminished clinical symptoms and ameliorated histological hallmarks of neuroinflammation. T cells in peripheral lymphoid organs of these mice produced less pro-inflammatory cytokines while their proliferation and the abundance of regulatory T cells were unaltered. The numbers of inflammatory monocytes and reactive microglia in the central nervous system (CNS) in MRlysM mice were significantly lower and they adopted an M2-polarized phenotype based on their gene expression profile, presumably explaining the ameliorated neuroinflammation. Our results indicate that the MR in myeloid cells plays a critical role for CNS autoimmunity, providing a rational to interfere with diseases such as MS by pharmacologically targeting this receptor.
Collapse
Affiliation(s)
- Elena Montes-Cobos
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Nils Schweingruber
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany.,Institute of Neuroimmunology, University Medical Center Goettingen, Goettingen, Germany.,Institute for Multiple Sclerosis Research, University Medical Center Goettingen, Goettingen, Germany
| | - Xiao Li
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Henrike J Fischer
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany.,Institute of Neuroimmunology, University Medical Center Goettingen, Goettingen, Germany.,Institute for Multiple Sclerosis Research, University Medical Center Goettingen, Goettingen, Germany
| | - Holger M Reichardt
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Fred Lühder
- Institute of Neuroimmunology, University Medical Center Goettingen, Goettingen, Germany.,Institute for Multiple Sclerosis Research, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
158
|
Menzel F, Kaiser N, Haehnel S, Rapp F, Patties I, Schöneberg N, Haimon Z, Immig K, Bechmann I. Impact of X-irradiation on microglia. Glia 2017; 66:15-33. [DOI: 10.1002/glia.23239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022]
Affiliation(s)
| | - Nicole Kaiser
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Susann Haehnel
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Felicitas Rapp
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Ina Patties
- Department of Radiation Therapy; Leipzig University; Leipzig Germany
| | | | - Zhana Haimon
- Department of Immunology; Weizmann Institute of Science; Rehovot Israel
| | - Kerstin Immig
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University; Leipzig Germany
| |
Collapse
|
159
|
Splitting the "Unsplittable": Dissecting Resident and Infiltrating Macrophages in Experimental Autoimmune Encephalomyelitis. Int J Mol Sci 2017; 18:ijms18102072. [PMID: 28961183 PMCID: PMC5666754 DOI: 10.3390/ijms18102072] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 09/23/2017] [Accepted: 09/25/2017] [Indexed: 12/12/2022] Open
Abstract
Macrophages predominate the inflammatory landscape within multiple sclerosis (MS) lesions, not only regarding cellularity but also with respect to the diverse functions this cell fraction provides during disease progression and remission. Researchers have been well aware of the fact that the macrophage pool during central nervous system (CNS) autoimmunity consists of a mixture of myeloid cells. Yet, separating these populations to define their unique contribution to disease pathology has long been challenging due to their similar marker expression. Sophisticated lineage tracing approaches as well as comprehensive transcriptome analysis have elevated our insight into macrophage biology to a new level enabling scientists to dissect the roles of resident (microglia and non-parenchymal macrophages) and infiltrating macrophages with unprecedented precision. To do so in an accurate way, researchers have to know their toolbox, which has been filled with diverse, discriminating approaches from decades of studying neuroinflammation in animal models. Every method has its own strengths and weaknesses, which will be addressed in this review. The focus will be on tools to manipulate and/or identify different macrophage subgroups within the injured murine CNS.
Collapse
|
160
|
De Feo D, Merlini A, Brambilla E, Ottoboni L, Laterza C, Menon R, Srinivasan S, Farina C, Garcia Manteiga JM, Butti E, Bacigaluppi M, Comi G, Greter M, Martino G. Neural precursor cell-secreted TGF-β2 redirects inflammatory monocyte-derived cells in CNS autoimmunity. J Clin Invest 2017; 127:3937-3953. [PMID: 28945200 DOI: 10.1172/jci92387] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 08/02/2017] [Indexed: 12/28/2022] Open
Abstract
In multiple sclerosis, the pathological interaction between autoreactive Th cells and mononuclear phagocytes in the CNS drives initiation and maintenance of chronic neuroinflammation. Here, we found that intrathecal transplantation of neural stem/precursor cells (NPCs) in mice with experimental autoimmune encephalomyelitis (EAE) impairs the accumulation of inflammatory monocyte-derived cells (MCs) in the CNS, leading to improved clinical outcome. Secretion of IL-23, IL-1, and TNF-α, the cytokines required for terminal differentiation of Th cells, decreased in the CNS of NPC-treated mice, consequently inhibiting the induction of GM-CSF-producing pathogenic Th cells. In vivo and in vitro transcriptome analyses showed that NPC-secreted factors inhibit MC differentiation and activation, favoring the switch toward an antiinflammatory phenotype. Tgfb2-/- NPCs transplanted into EAE mice were ineffective in impairing MC accumulation within the CNS and failed to drive clinical improvement. Moreover, intrathecal delivery of TGF-β2 during the effector phase of EAE ameliorated disease severity. Taken together, these observations identify TGF-β2 as the crucial mediator of NPC immunomodulation. This study provides evidence that intrathecally transplanted NPCs interfere with the CNS-restricted inflammation of EAE by reprogramming infiltrating MCs into antiinflammatory myeloid cells via secretion of TGF-β2.
Collapse
Affiliation(s)
| | | | | | | | | | - Ramesh Menon
- Immunobiology of Neurological Disorders Lab, Institute of Experimental Neurology, Division of Neuroscience, and
| | - Sundararajan Srinivasan
- Immunobiology of Neurological Disorders Lab, Institute of Experimental Neurology, Division of Neuroscience, and
| | - Cinthia Farina
- Immunobiology of Neurological Disorders Lab, Institute of Experimental Neurology, Division of Neuroscience, and
| | - Jose Manuel Garcia Manteiga
- Center for Translational Genomics and BioInformatics, San Raffaele Scientific Institute and Vita Salute San Raffaele University, Milan, Italy
| | | | | | | | - Melanie Greter
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | | |
Collapse
|
161
|
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: 341] [Impact Index Per Article: 48.7] [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
|
162
|
Liu J, Xue Y, Dong D, Xiao C, Lin C, Wang H, Song F, Fu T, Wang Z, Chen J, Pan H, Li Y, Cai D, Li Z. CCR2 - and CCR2 + corneal macrophages exhibit distinct characteristics and balance inflammatory responses after epithelial abrasion. Mucosal Immunol 2017; 10:1145-1159. [PMID: 28120849 PMCID: PMC5562841 DOI: 10.1038/mi.2016.139] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/19/2016] [Indexed: 02/04/2023]
Abstract
Macrophages are distributed throughout the body and are crucial for the restoration of damaged tissues. However, their characteristics in the cornea and roles in the repair of corneal injures are unclear. Here we show that corneal macrophages can be classified as CCR2- macrophages, which already exist in the cornea at embryonic day 12.5 (E12.5) and are similar to yolk sac-derived macrophages, microglia, in phenotype and gene expression, and CCR2+ macrophages, which do not appear in the cornea until E17.5. At a steady state, CCR2- corneal macrophages have local proliferation capacity and are rarely affected by monocytes; however, following corneal epithelial abrasion, most CCR2- corneal macrophages are replaced by monocytes. In contrast, CCR2+ macrophages are repopulated by monocytes under both a steady-state condition and following corneal wounding. Depletion of CCR2+ macrophages decreases corneal inflammation after epithelial abrasion, whereas depletion of CCR2- macrophages increases inflammation of the injured cornea. Loss of either cell type results in a delay in corneal healing. These data indicate that there are two unique macrophage populations present in the cornea, both of which participate in corneal wound healing by balancing the inflammatory response.
Collapse
Affiliation(s)
- J Liu
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China,International Ocular Surface Research Center and Institute of Ophthalmology, Jinan University Medical School, Guangzhou, China,Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Y Xue
- International Ocular Surface Research Center and Institute of Ophthalmology, Jinan University Medical School, Guangzhou, China
| | - D Dong
- International Ocular Surface Research Center and Institute of Ophthalmology, Jinan University Medical School, Guangzhou, China
| | - C Xiao
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - C Lin
- International Ocular Surface Research Center and Institute of Ophthalmology, Jinan University Medical School, Guangzhou, China
| | - H Wang
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - F Song
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - T Fu
- International Ocular Surface Research Center and Institute of Ophthalmology, Jinan University Medical School, Guangzhou, China
| | - Z Wang
- Department of Medical Images, The Third People’s Hospital, Puyang, China
| | - J Chen
- International Ocular Surface Research Center and Institute of Ophthalmology, Jinan University Medical School, Guangzhou, China
| | - H Pan
- International Ocular Surface Research Center and Institute of Ophthalmology, Jinan University Medical School, Guangzhou, China
| | - Y Li
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - D Cai
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Z Li
- International Ocular Surface Research Center and Institute of Ophthalmology, Jinan University Medical School, Guangzhou, China,Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China,Section of Leukocyte Biology, Department of Pediatrics, Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA,()
| |
Collapse
|
163
|
Abstract
Monocytes are short-lived mononuclear phagocytes that circulate in the bloodstream and comprise two main subpopulations that in the mouse are best defined by the Ly6C marker. Intravascular functions of "classical" Ly6C+ monocytes and their interactions with other lymphoid and myeloid leukocytes in the circulation remain poorly understood. Rather, these cells are known to efficiently extravasate into tissues. Indeed, Ly6C+ monocytes and their descendants have emerged as a third, highly plastic and dynamic cellular system that complements the two classical, tissue-resident mononuclear phagocyte compartments, i.e., macrophages and dendritic cells, on demand. Following recruitment to injured tissue, Ly6C+ monocytes respond to local cues and can critically contribute to the initiation and resolution of inflammatory reactions. The second main murine monocyte subset, Ly6C- cells, derive in steady state from Ly6C+ monocytes and remain in the vasculature, where the cells act as scavengers. Moreover, a major fraction of Ly6C- monocytes adheres to the capillary endothelium and patrols the vessel wall for surveillance. Given the central role of monocytes in homeostasis and pathology, in-depth study of this cellular compartment can be highly informative on the health state of the organism and provides an attractive target for therapeutic intervention.
Collapse
|
164
|
Konishi H, Kobayashi M, Kunisawa T, Imai K, Sayo A, Malissen B, Crocker PR, Sato K, Kiyama H. Siglec-H is a microglia-specific marker that discriminates microglia from CNS-associated macrophages and CNS-infiltrating monocytes. Glia 2017; 65:1927-1943. [PMID: 28836308 DOI: 10.1002/glia.23204] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/25/2017] [Accepted: 07/25/2017] [Indexed: 01/06/2023]
Abstract
Several types of myeloid cell are resident in the CNS. In the steady state, microglia are present in the CNS parenchyma, whereas macrophages reside in boundary regions of the CNS, such as perivascular spaces, the meninges and choroid plexus. In addition, monocytes infiltrate into the CNS parenchyma from circulation upon blood-brain barrier breakdown after CNS injury and inflammation. Although several markers, such as CD11b and ionized calcium-binding adapter molecule 1 (Iba1), are frequently used as microglial markers, they are also expressed by other types of myeloid cell and microglia-specific markers were not defined until recently. Previous transcriptome analyses of isolated microglia identified a transmembrane lectin, sialic acid-binding immunoglobulin-like lectin H (Siglec-H), as a molecular signature for microglia; however, this was not confirmed by histological studies in the nervous system and the reliability of Siglec-H as a microglial marker remained unclear. Here, we demonstrate that Siglec-H is an authentic marker for microglia in mice by immunohistochemistry using a Siglec-H-specific antibody. Siglec-H was expressed by parenchymal microglia from developmental stages to adulthood, and the expression was maintained in activated microglia under injury or inflammatory condition. However, Siglec-H expression was absent from CNS-associated macrophages and CNS-infiltrating monocytes, except for a minor subset of cells. We also show that the Siglech gene locus is a feasible site for specific targeting of microglia in the nervous system. In conclusion, Siglec-H is a reliable marker for microglia that will allow histological identification of microglia and microglia-specific gene manipulation in the nervous system.
Collapse
Affiliation(s)
- Hiroyuki Konishi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Masaaki Kobayashi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Taikan Kunisawa
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Kenta Imai
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Akira Sayo
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.,Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS UMR, Marseille, 13288, France
| | - Paul R Crocker
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Katsuaki Sato
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, 889-1692, Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| |
Collapse
|
165
|
Foxp3-independent mechanism by which TGF-β controls peripheral T cell tolerance. Proc Natl Acad Sci U S A 2017; 114:E7536-E7544. [PMID: 28827353 DOI: 10.1073/pnas.1706356114] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Peripheral T cell tolerance is promoted by the regulatory cytokine TGF-β and Foxp3-expressing Treg cells. However, whether TGF-β and Treg cells are part of the same regulatory module, or exist largely as distinct pathways to repress self-reactive T cells remains incompletely understood. Using a transgenic model of autoimmune diabetes, here we show that ablation of TGF-β receptor II (TβRII) in T cells, but not Foxp3 deficiency, resulted in early-onset diabetes with complete penetrance. The rampant autoimmune disease was associated with enhanced T cell priming and elevated T cell expression of the inflammatory cytokine GM-CSF, concomitant with pancreatic infiltration of inflammatory monocytes that triggered immunopathology. Ablation of the GM-CSF receptor alleviated the monocyte response and inhibited disease development. These findings reveal that TGF-β promotes T cell tolerance primarily via Foxp3-independent mechanisms and prevents autoimmunity in this model by repressing the cross talk between adaptive and innate immune systems.
Collapse
|
166
|
Lyck R, Lécuyer MA, Abadier M, Wyss CB, Matti C, Rosito M, Enzmann G, Zeis T, Michel L, García Martín AB, Sallusto F, Gosselet F, Deutsch U, Weiner JA, Schaeren-Wiemers N, Prat A, Engelhardt B. ALCAM (CD166) is involved in extravasation of monocytes rather than T cells across the blood-brain barrier. J Cereb Blood Flow Metab 2017; 37:2894-2909. [PMID: 28273717 PMCID: PMC5536797 DOI: 10.1177/0271678x16678639] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Activated leukocyte cell adhesion molecule (ALCAM) has been proposed to mediate leukocyte migration across the blood-brain barrier (BBB) in multiple sclerosis or experimental autoimmune encephalomyelitis (EAE). Here, we confirmed vascular ALCAM expression in human brain tissue samples in situ and on two different human in vitro BBB models. Antibody-mediated inhibition of ALCAM reduced diapedesis of human CD4+ Th1 but not of Th17 cells across the human BBB in vitro. In accordance to human Th1 cells, mouse Th1 cells showed reduced diapedesis across an ALCAM-/- in vitro BBB model under static but no longer under flow conditions. In contrast to the limited role of ALCAM in T cell extravasation across the BBB, we found a contribution of ALCAM to rolling, adhesion, and diapedesis of human CD14+ monocytes across the human BBB under flow and static conditions. Taken together, our study highlights the potential differences in the CNS expression of ALCAM in mouse and human and supports a prominent role for ALCAM in the multi-step extravasation of monocytes across the BBB.
Collapse
Affiliation(s)
- Ruth Lyck
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Marc-André Lécuyer
- 2 Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Neuroimmunology Research Laboratory, Montréal, Québec, Canada
| | - Michael Abadier
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Christof B Wyss
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Christoph Matti
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Maria Rosito
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gaby Enzmann
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Thomas Zeis
- 3 Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Laure Michel
- 2 Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Neuroimmunology Research Laboratory, Montréal, Québec, Canada
| | | | | | | | - Urban Deutsch
- 1 Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Joshua A Weiner
- 6 Departments of Biology and Psychiatry, The University of Iowa, Iowa City, IA, USA
| | - Nicole Schaeren-Wiemers
- 3 Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexandre Prat
- 2 Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Neuroimmunology Research Laboratory, Montréal, Québec, Canada
| | | |
Collapse
|
167
|
Lund H, Pieber M, Harris RA. Lessons Learned about Neurodegeneration from Microglia and Monocyte Depletion Studies. Front Aging Neurosci 2017; 9:234. [PMID: 28804456 PMCID: PMC5532389 DOI: 10.3389/fnagi.2017.00234] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/05/2017] [Indexed: 12/20/2022] Open
Abstract
While bone marrow-derived Ly6Chi monocytes can infiltrate the central nervous system (CNS) they are developmentally and functionally distinct from resident microglia. Our understanding of the relative importance of these two populations in the distinct processes of pathogenesis and resolution of inflammation during neurodegenerative disorders was limited by a lack of tools to specifically manipulate each cell type. During recent years, the development of experimental cell-specific depletion models has enabled this issue to be addressed. Herein we compare and contrast the different depletion approaches that have been used, focusing on the respective functionalities of microglia and monocyte-derived macrophages in a range of neurodegenerative disease states, and discuss their prospects for immunotherapy.
Collapse
Affiliation(s)
- Harald Lund
- Department of Clinical Neuroscience, Karolinska Institutet, Centre for Molecular Medicine, Karolinska Hospital at SolnaSolna, Sweden
| | - Melanie Pieber
- Department of Clinical Neuroscience, Karolinska Institutet, Centre for Molecular Medicine, Karolinska Hospital at SolnaSolna, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Centre for Molecular Medicine, Karolinska Hospital at SolnaSolna, Sweden
| |
Collapse
|
168
|
Acetylcholine-producing NK cells attenuate CNS inflammation via modulation of infiltrating monocytes/macrophages. Proc Natl Acad Sci U S A 2017; 114:E6202-E6211. [PMID: 28696300 DOI: 10.1073/pnas.1705491114] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The nonneural cholinergic system of immune cells is pivotal for the maintenance of immunological homeostasis. Here we demonstrate the expression of choline acetyltransferase (ChAT) and cholinergic enzymes in murine natural killer (NK) cells. The capacity for acetylcholine synthesis by NK cells increased markedly under inflammatory conditions such as experimental autoimmune encephalomyelitis (EAE), in which ChAT expression escalated along with the maturation of NK cells. ChAT+ and ChAT- NK cells displayed distinctive features in terms of cytotoxicity and chemokine/cytokine production. Transfer of ChAT+ NK cells into the cerebral ventricles of CX3CR1-/- mice reduced brain and spinal cord damage after EAE induction, and decreased the numbers of CNS-infiltrating CCR2+Ly6Chi monocytes. ChAT+ NK cells killed CCR2+Ly6Chi monocytes directly via the disruption of tolerance and inhibited the production of proinflammatory cytokines. Interestingly, ChAT+ NK cells and CCR2+Ly6Chi monocytes formed immune synapses; moreover, the impact of ChAT+ NK cells was mediated by α7-nicotinic acetylcholine receptors. Finally, the NK cell cholinergic system up-regulated in response to autoimmune activation in multiple sclerosis, perhaps reflecting the severity of disease. Therefore, this study extends our understanding of the nonneural cholinergic system and the protective immune effect of acetylcholine-producing NK cells in autoimmune diseases.
Collapse
|
169
|
Abstract
STUDY DESIGN Prospective observational study. OBJECTIVES To describe the correlation between CCL-2, CCL-3, CCL-4 and CXCL-5 serum levels and remission after traumatic spinal cord injury (SCI) in a human protocol compared with animal studies. SETTING Germany, Rhineland-Palatinate (Rheinland-Pfalz). METHODS We examined the serum levels of CCL-2, CCL-3, CCL-4 and CXCL-5 over a 12-week period; in particular, at admission and 4, 9 and 12 h, 1 and 3 days and 1, 2, 4, 8 and 12 weeks after trauma. According to our study design, we matched 10 patients with TSCI and neurological remission with 10 patients with an initial ASIA A grade and no neurological remission. In all, 10 patients with vertebral fracture without neurological deficits served as control. Our analysis was performed using a Luminex Cytokine Panel. Multivariate logistic regression models were used to examine the predictive value with respect to neurological remission vs no neurological remission. RESULTS The results of our study showed differences in the serum expression patterns of CCL-2 in association with the neurological remission (CCL-2 at admission P=0.013). Serum levels of CCL-2 and CCL-4 were significantly different in patients with and without neurological remission. The favored predictive model resulted in an area under the curve (AUC) of 93.1% in the receiver operating characteristic (ROC) analysis. CONCLUSIONS Our results indicate that peripheral serum analysis is a suitable concept for predicting the patient's potential for neurological remission after TSCI. Furthermore, the initial CCL-2 concentration provides an additional predictive value compared with the NLI (neurological level of injury). Therefore, the present study introduces a promising approach for future monitoring concepts and tracking techniques for current therapies. The results indicate that future investigations with an enlarged sample size are needed in order to develop monitoring, prognostic and scoring systems.
Collapse
|
170
|
Herz J, Filiano AJ, Wiltbank AT, Yogev N, Kipnis J. Myeloid Cells in the Central Nervous System. Immunity 2017; 46:943-956. [PMID: 28636961 PMCID: PMC5657250 DOI: 10.1016/j.immuni.2017.06.007] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/17/2017] [Accepted: 06/02/2017] [Indexed: 02/07/2023]
Abstract
The central nervous system (CNS) and its meningeal coverings accommodate a diverse myeloid compartment that includes parenchymal microglia and perivascular macrophages, as well as choroid plexus and meningeal macrophages, dendritic cells, and granulocytes. These myeloid populations enjoy an intimate relationship with the CNS, where they play an essential role in both health and disease. Although the importance of these cells is clearly recognized, their exact function in the CNS continues to be explored. Here, we review the subsets of myeloid cells that inhabit the parenchyma, meninges, and choroid plexus and discuss their roles in CNS homeostasis. We also discuss the role of these cells in various neurological pathologies, such as autoimmunity, mechanical injury, neurodegeneration, and infection. We highlight the neuroprotective nature of certain myeloid cells by emphasizing their therapeutic potential for the treatment of neurological conditions.
Collapse
Affiliation(s)
- Jasmin Herz
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Anthony J Filiano
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | - Ashtyn T Wiltbank
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Nir Yogev
- Gutenberg Research Fellowship Group of Neuroimmunology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Gutenberg Research Fellowship Group of Neuroimmunology, Focus Program Translational Neuroscience and Immunotherapy, Rhine Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| |
Collapse
|
171
|
Tumor Necrosis Factor Alpha-Induced Recruitment of Inflammatory Mononuclear Cells Leads to Inflammation and Altered Brain Development in Murine Cytomegalovirus-Infected Newborn Mice. J Virol 2017; 91:JVI.01983-16. [PMID: 28122986 PMCID: PMC5375689 DOI: 10.1128/jvi.01983-16] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/06/2017] [Indexed: 12/24/2022] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection is a significant cause of abnormal neurodevelopment and long-term neurological sequelae in infants and children. Resident cell populations of the developing brain have been suggested to be more susceptible to virus-induced cytopathology, a pathway thought to contribute to the clinical outcomes following intrauterine HCMV infection. However, recent findings in a newborn mouse model of the infection in the developing brain have indicated that elevated levels of proinflammatory mediators leading to mononuclear cell activation and recruitment could underlie the abnormal neurodevelopment. In this study, we demonstrate that treatment with tumor necrosis factor alpha (TNF-α)-neutralizing antibodies decreased the frequency of CD45+ Ly6Chi CD11b+ CCR2+ activated myeloid mononuclear cells (MMCs) and the levels of proinflammatory cytokines in the blood and the brains of murine CMV-infected mice. This treatment also normalized neurodevelopment in infected mice without significantly impacting the level of virus replication. These results indicate that TNF-α is a major component of the inflammatory response associated with altered neurodevelopment that follows murine CMV infection of the developing brain and that a subset of peripheral blood myeloid mononuclear cells represent a key effector cell population in this model of virus-induced inflammatory disease of the developing brain.IMPORTANCE Congenital human cytomegalovirus (HCMV) infection is the most common viral infection of the developing human fetus and can result in neurodevelopmental sequelae. Mechanisms of disease leading to neurodevelopmental deficits in infected infants remain undefined, but postulated pathways include loss of neuronal progenitor cells, damage to the developing vascular system of the brain, and altered cellular positioning. Direct virus-mediated cytopathic effects cannot explain the phenotypes of brain damage in most infected infants. Using a mouse model that recapitulates characteristics of the brain infection described in human infants, we have shown that TNF-α plays a key role in brain inflammation, including recruitment of inflammatory mononuclear cells. Neutralization of TNF-α normalized neurodevelopmental abnormalities in infected mice, providing evidence that virus-induced inflammation is a major component of disease in the developing brain. These results suggest that interventions limiting inflammation associated with the infection could potentially improve the neurologic outcome of infants infected in utero with HCMV.
Collapse
|
172
|
Wolf Y, Shemer A, Polonsky M, Gross M, Mildner A, Yona S, David E, Kim KW, Goldmann T, Amit I, Heikenwalder M, Nedospasov S, Prinz M, Friedman N, Jung S. Autonomous TNF is critical for in vivo monocyte survival in steady state and inflammation. J Exp Med 2017; 214:905-917. [PMID: 28330904 PMCID: PMC5379969 DOI: 10.1084/jem.20160499] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 12/30/2016] [Accepted: 02/15/2017] [Indexed: 11/04/2022] Open
Abstract
Monocytes are circulating mononuclear phagocytes, poised to extravasate to sites of inflammation and differentiate into monocyte-derived macrophages and dendritic cells. Tumor necrosis factor (TNF) and its receptors are up-regulated during monopoiesis and expressed by circulating monocytes, as well as effector monocytes infiltrating certain sites of inflammation, such as the spinal cord, during experimental autoimmune encephalomyelitis (EAE). In this study, using competitive in vitro and in vivo assays, we show that monocytes deficient for TNF or TNF receptors are outcompeted by their wild-type counterpart. Moreover, monocyte-autonomous TNF is critical for the function of these cells, as TNF ablation in monocytes/macrophages, but not in microglia, delayed the onset of EAE in challenged animals and was associated with reduced acute spinal cord infiltration of Ly6Chi effector monocytes. Collectively, our data reveal a previously unappreciated critical cell-autonomous role of TNF on monocytes for their survival, maintenance, and function.
Collapse
Affiliation(s)
- Yochai Wolf
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Anat Shemer
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Michal Polonsky
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Mor Gross
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander Mildner
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Simon Yona
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ki-Wook Kim
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tobias Goldmann
- Institute for Neuropathology, University of Freiburg, 79085 Freiburg, Germany
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Mathias Heikenwalder
- Institut für Virologie, Helmholtz Zentrum München, 85764 Neuherberg, Germany.,Department of Chronic Inflammation and Cancer, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Sergei Nedospasov
- Engelhardt Institute of Molecular Biology, Moscow, Russia 119991.,German Rheumatism Research Center, 10117 Berlin, Germany
| | - Marco Prinz
- Institute for Neuropathology, University of Freiburg, 79085 Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79085 Freiburg, Germany
| | - Nir Friedman
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
173
|
Menasria R, Canivet C, Piret J, Gosselin J, Boivin G. Protective role of CX3CR1 signalling in resident cells of the central nervous system during experimental herpes simplex virus encephalitis. J Gen Virol 2017; 98:447-460. [PMID: 27902351 DOI: 10.1099/jgv.0.000667] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
CX3CR1 is an important chemokine receptor expressed on the surface of microglia and blood leukocytes, including monocytes. Signalling through this receptor influences the immune activity of microglia and monocyte trafficking into the central nervous system (CNS) in several neurological diseases. During experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE), CX3CR1 deficiency has been reported to exacerbate the outcome of the disease. However, the precise contribution of CX3CR1 expressed in resident cells of the CNS or peripheral monocytes in protection against HSE remains unclear. To dissect the role of CX3CR1 during HSE, we reconstituted irradiated C57BL/6 WT and CX3CR1-/- mice with CX3CR1-/- (CX3CR1-/-→WT) and WT (WT→CX3CR1-/-) bone marrow cells, respectively. Our results showed that following intranasal infection with 1.2×106 p.f.u. of HSV-1, mortality rates were significantly higher in CX3CR1-/- (61.7 %) and WT→CX3CR1-/- (66.2 %) compared to WT (16.6 %; P=0.012 and P=0.016, respectively) and CX3CR1-/-→WT animals (20 %; P=0.013 and P=0.011, respectively). Higher mortality rates in CX3CR1-/- and WT→CX3CR1-/- mice were associated with increased infectious viral titres and wider HSV dissemination in brains, as well as an overproduction of inflammatory cytokines and chemokines including IL-1β, IL-6, IFN-γ, C-C motif ligand 2 and C-C motif ligand 5. Furthermore, CX3CR1 deficiency in resident cells of the CNS resulted in excessive and sustained Ly6Chi inflammatory monocyte and neutrophil infiltration into the brain. These data suggest that CX3CR1 deficiency in resident cells of the CNS affects mouse survival, HSV-1 replication control and cerebral inflammatory response whereas its deficiency in the haematopoietic system does not appear to influence the outcome of HSE.
Collapse
Affiliation(s)
- Rafik Menasria
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| | - Coraline Canivet
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| | - Jocelyne Piret
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| | - Jean Gosselin
- Laboratory of Innate Immunology of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases of the CHU of Quebec and Laval University, 2705 Boul Laurier, Quebec City, QC G1V 4G2, Canada
| |
Collapse
|
174
|
Wegner A, Verhagen J, Wraith DC. Myeloid-derived suppressor cells mediate tolerance induction in autoimmune disease. Immunology 2017; 151:26-42. [PMID: 28140447 DOI: 10.1111/imm.12718] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 01/05/2023] Open
Abstract
In multiple sclerosis (MS) T cells aberrantly recognize self-peptides of the myelin sheath and attack the central nervous system (CNS). Antigen-specific peptide immunotherapy, which aims to restore tolerance while avoiding the use of non-specific immunosuppressive drugs, is a promising approach to combat autoimmune disease, but the cellular mechanisms behind successful therapy remain poorly understood. Myeloid-derived suppressor cells (MDSCs) have been studied intensively in the field of cancer and to a lesser extent in autoimmunity. Because of their suppressive effect on the immune system in cancer, we hypothesized that the development of MDSCs and their interaction with CD4+ T cells could be beneficial for antigen-specific immunotherapy. Hence, changes in the quantity, phenotype and function of MDSCs during tolerance induction in our model of MS were evaluated. We reveal, for the first time, an involvement of a subset of MDSCs, known as polymorphonuclear (PMN)-MDSCs, in the process of tolerance induction. PMN-MDSCs were shown to adopt a more suppressive phenotype during peptide immunotherapy and inhibit CD4+ T-cell proliferation in a cell-contact-dependent manner, mediated by arginase-1. Moreover, increased numbers of tolerogenic PMN-MDSCs, such as observed over the course of peptide immunotherapy, were demonstrated to provide protection from disease in a model of experimental autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- Anja Wegner
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Johan Verhagen
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - David C Wraith
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
175
|
Sphingosine 1-phosphate receptor modulation suppresses pathogenic astrocyte activation and chronic progressive CNS inflammation. Proc Natl Acad Sci U S A 2017; 114:2012-2017. [PMID: 28167760 DOI: 10.1073/pnas.1615413114] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of the CNS that causes disability in young adults as a result of the irreversible accumulation of neurological deficits. Although there are potent disease-modifying agents for its initial relapsing-remitting phase, these therapies show limited efficacy in secondary progressive MS (SPMS). Thus, there is an unmet clinical need for the identification of disease mechanisms and potential therapeutic approaches for SPMS. Here, we show that the sphingosine 1-phosphate receptor (S1PR) modulator fingolimod (FTY720) ameliorated chronic progressive experimental autoimmune encephalomyelitis in nonobese diabetic mice, an experimental model that resembles several aspects of SPMS, including neurodegeneration and disease progression driven by the innate immune response in the CNS. Indeed, S1PR modulation by FTY720 in murine and human astrocytes suppressed neurodegeneration-promoting mechanisms mediated by astrocytes, microglia, and CNS-infiltrating proinflammatory monocytes. Genome-wide studies showed that FTY720 suppresses transcriptional programs associated with the promotion of disease progression by astrocytes. The study of the molecular mechanisms controlling these transcriptional modules may open new avenues for the development of therapeutic strategies for progressive MS.
Collapse
|
176
|
The role of peripheral immune cells in the CNS in steady state and disease. Nat Neurosci 2017; 20:136-144. [DOI: 10.1038/nn.4475] [Citation(s) in RCA: 336] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/08/2016] [Indexed: 02/07/2023]
|
177
|
Mildner A. Ghosts in the shell: identification of microglia in the human central nervous system by P2Y12 receptor. Neural Regen Res 2017; 12:570-571. [PMID: 28553330 PMCID: PMC5436348 DOI: 10.4103/1673-5374.205090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
178
|
Barnie PA, Zhang P, Lv H, Wang D, Su X, Su Z, Xu H. Myeloid-derived suppressor cells and myeloid regulatory cells in cancer and autoimmune disorders. Exp Ther Med 2016; 13:378-388. [PMID: 28352304 DOI: 10.3892/etm.2016.4018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/17/2016] [Indexed: 12/19/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) were originally described as a heterogeneous population of immature cells derived from myeloid progenitors with immune-suppressive functions in tumor-bearing hosts. In recent years, increasing number of studies have described various populations of myeloid cells with MDSC-like properties in murine models of cancer and autoimmune diseases. These studies have observed that the populations of MDSCs are increased during inflammation and autoimmune conditions. In addition, MDSCs can effectively suppress T cell responses and modulate the activity of natural killer cells and other myeloid cells. MDSCs have also been implicated in the induction of regulatory T cell production. Furthermore, these cells have the potential to suppress the autoimmune response, thereby limiting tissue injury. Myeloid regulatory cells (Mregs) are recently attracting increasing attention, since they function in proinflammatory and immune suppression in autoimmune diseases, as well as in various types of cancer. Currently, research focus is directed from MDSCs to Mregs in cancer and autoimmune diseases. The present study reviewed the suppressive roles of MDSCs in various autoimmune murine models, the immune modulation of MDSCs to T helper 17 lymphocytes, as well as the proinflammatory and immunosuppressive roles of Mregs in various types of cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Prince Amoah Barnie
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China; Department of Biomedical and Forensic Sciences, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Pan Zhang
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hongxiang Lv
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Dan Wang
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiaolian Su
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhaoliang Su
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China; Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P.R. China
| | - Huaxi Xu
- Department of Immunology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
179
|
Menasria R, Canivet C, Piret J, Gosselin J, Boivin G. Both Cerebral and Hematopoietic Deficiencies in CCR2 Result in Uncontrolled Herpes Simplex Virus Infection of the Central Nervous System in Mice. PLoS One 2016; 11:e0168034. [PMID: 27930721 PMCID: PMC5145225 DOI: 10.1371/journal.pone.0168034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/23/2016] [Indexed: 12/11/2022] Open
Abstract
CCR2 is a chemokine receptor expressed on the surface of blood leukocytes, particularly «Ly6Chi» inflammatory monocytes and microglia. Signaling through this receptor is thought to influence the immune activity of microglia as well as monocytes egress from the bone marrow (BM) and their trafficking into the central nervous system (CNS) in several neurological diseases. During experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE), CCR2 deficiency has been reported to exacerbate the outcome of the disease. However, the precise contribution of CCR2 expressed in cells of the CNS or peripheral monocytes in the protection against HSE remains unclear. To dissect the differential role of CCR2 during HSE, chimeric mice with receptor deficiency in the brain or blood cells were generated by transplanting wild-type (WT) C57BL/6 or CCR2-/- BM-derived cells in CCR2-/- (WT→CCR2-/-) and WT (CCR2-/-→WT) mice, respectively. Our results indicate that following intranasal infection with 1.2x106 plaque forming units of HSV-1, CCR2 deficiency in hematopoietic cells and, to a lesser extent, in CNS exacerbates the outcome of HSE. Mortality rates of CCR2-/- (71.4%) and CCR2-/-→WT (57.1%) mice were significantly higher than that of WT (15.3%; P<0.01 and P<0.05, respectively) but the difference did not reach statistical significance for WT→CCR2-/- animals (42.8%; P = 0.16). Both peripheral and CNS deficiencies in CCR2 resulted in increased infectious viral titers and wider dissemination of HSV antigens in the brain as well as an overproduction of inflammatory cytokines and chemokines including IL-1β, IL-6, CCL2, CCL3 and CCL5. Furthermore, CCR2 deficiency in the hematopoietic system altered monocytes egress from the BM and their recruitment to the CNS, which may contribute to the failure in HSV-1 containment. Collectively, these data suggest that CCR2 expressed on cells of CNS and especially on peripheral monocytes is important for the control of HSV-1 replication and inflammatory environment during experimental HSE.
Collapse
Affiliation(s)
- Rafik Menasria
- Research Center in Infectious Diseases, CHU of Quebec Research Center, Department of microbiology-immunology and infectious diseases, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Coraline Canivet
- Research Center in Infectious Diseases, CHU of Quebec Research Center, Department of microbiology-immunology and infectious diseases, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Jocelyne Piret
- Research Center in Infectious Diseases, CHU of Quebec Research Center, Department of microbiology-immunology and infectious diseases, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Jean Gosselin
- Laboratory of Innate Immunity, CHU of Quebec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Guy Boivin
- Research Center in Infectious Diseases, CHU of Quebec Research Center, Department of microbiology-immunology and infectious diseases, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| |
Collapse
|
180
|
Abstract
Cytokines provide cells with the ability to communicate with one another and orchestrate complex multicellular behaviour. There is an emerging understanding of the role that cytokines play in normal homeostatic tissue function and how dysregulation of these cytokine networks is associated with pathological conditions. The central nervous system (CNS), where few blood-borne immune cells circulate, seems to be particularly vulnerable to dysregulated cytokine networks. In degenerative diseases, such as proteopathies, CNS-resident cells are the predominant producers of pro-inflammatory cytokines. By contrast, in classical neuroinflammatory diseases, such as multiple sclerosis and encephalitides, pro-inflammatory cytokines are mainly produced by tissue-invading leukocytes. Whereas the effect of dysregulated cytokine networks in proteopathies is controversial, cytokines delivered to the CNS by invading immune cells are in general detrimental to the tissue. Here, we summarize recent observations on the impact of dysregulated cytokine networks in neuroinflammation.
Collapse
|
181
|
Harrison-Brown M, Liu GJ, Banati R. Checkpoints to the Brain: Directing Myeloid Cell Migration to the Central Nervous System. Int J Mol Sci 2016; 17:E2030. [PMID: 27918464 PMCID: PMC5187830 DOI: 10.3390/ijms17122030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/23/2016] [Accepted: 11/25/2016] [Indexed: 12/13/2022] Open
Abstract
Myeloid cells are a unique subset of leukocytes with a diverse array of functions within the central nervous system during health and disease. Advances in understanding of the unique properties of these cells have inspired interest in their use as delivery vehicles for therapeutic genes, proteins, and drugs, or as "assistants" in the clean-up of aggregated proteins and other molecules when existing drainage systems are no longer adequate. The trafficking of myeloid cells from the periphery to the central nervous system is subject to complex cellular and molecular controls with several 'checkpoints' from the blood to their destination in the brain parenchyma. As important components of the neurovascular unit, the functional state changes associated with lineage heterogeneity of myeloid cells are increasingly recognized as important for disease progression. In this review, we discuss some of the cellular elements associated with formation and function of the neurovascular unit, and present an update on the impact of myeloid cells on central nervous system (CNS) diseases in the laboratory and the clinic. We then discuss emerging strategies for harnessing the potential of site-directed myeloid cell homing to the CNS, and identify promising avenues for future research, with particular emphasis on the importance of untangling the functional heterogeneity within existing myeloid subsets.
Collapse
Affiliation(s)
- Meredith Harrison-Brown
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2141, Australia.
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia.
| | - Guo-Jun Liu
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2141, Australia.
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia.
| | - Richard Banati
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2141, Australia.
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia.
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
182
|
Zhou Z, Ding M, Huang L, Gilkeson G, Lang R, Jiang W. Toll-like receptor-mediated immune responses in intestinal macrophages; implications for mucosal immunity and autoimmune diseases. Clin Immunol 2016; 173:81-86. [PMID: 27620642 PMCID: PMC5148676 DOI: 10.1016/j.clim.2016.09.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/04/2016] [Accepted: 09/07/2016] [Indexed: 12/21/2022]
Abstract
Monocytes are precursors of macrophages and key players during inflammation and pathogen challenge in the periphery, whereas intestinal resident macrophages act as innate effector cells to engulf and clear bacteria, secrete cytokines, and maintain intestinal immunity and homeostasis. However, perturbation of toll-like receptor signaling pathway in intestinal macrophages has been associated with tolerance breakdown in autoimmune diseases. In the present review, we have summarized and discussed the role of toll-like receptor signals in human intestinal macrophages, and the role of human intestinal macrophages in keeping human intestinal immunity, homeostasis, and autoimmune diseases.
Collapse
Affiliation(s)
- Zejun Zhou
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston 29425, USA
| | - Miao Ding
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston 29425, USA
| | - Lei Huang
- The 302 Hospital of PLA, Treatment and Research Center for Infectious Diseases, Beijing 100039, China
| | - Gary Gilkeson
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston 29425, SC, USA
| | - Ren Lang
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 10020, China.
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston 29425, USA; Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston 29425, USA.
| |
Collapse
|
183
|
Sie C, Korn T. Dendritic cells in central nervous system autoimmunity. Semin Immunopathol 2016; 39:99-111. [PMID: 27888330 DOI: 10.1007/s00281-016-0608-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/13/2016] [Indexed: 02/01/2023]
Abstract
Dendritic cells (DCs) operate at the intersection of the innate and adaptive immune systems. DCs can promote or inhibit adaptive immune responses against neuroantigens. While DC intrinsic properties, i.e., their maturation state or the subset they belong to, are important determinants of the outcome of an autoimmune reaction, tissue-specific cues might also be relevant for the function of DCs. Thus, a better understanding of the performance of distinct DC subsets in specific anatomical niches, not only in lymphoid tissue but also in non-lymphoid tissues such as the meninges, the choroid plexus, and the inflamed CNS parenchyma, will be instrumental for the design of immune intervention strategies to chronic inflammatory diseases that do not put at risk basic surveillance functions of the immune system in the CNS. Here, we will review modern concepts of DC biology in steady state and during autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Christopher Sie
- Klinikum rechts der Isar, Department of Neurology and Department of Experimental Neuroimmunology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Thomas Korn
- Klinikum rechts der Isar, Department of Neurology and Department of Experimental Neuroimmunology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
184
|
Mildner A, Huang H, Radke J, Stenzel W, Priller J. P2Y12receptor is expressed on human microglia under physiological conditions throughout development and is sensitive to neuroinflammatory diseases. Glia 2016; 65:375-387. [DOI: 10.1002/glia.23097] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 10/18/2016] [Accepted: 10/24/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Alexander Mildner
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry; Charité-Universitätsmedizin Berlin; Berlin 10117 Germany
| | - Hao Huang
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry; Charité-Universitätsmedizin Berlin; Berlin 10117 Germany
| | - Josefine Radke
- Department of Neuropathology; Charité-Universitätsmedizin Berlin; Berlin 10117 Germany
- Berlin Institute of Health (BIH); Berlin Germany
- German Consortium for Translational Cancer Research (DKTK); Heidelberg Germany
| | - Werner Stenzel
- Department of Neuropathology; Charité-Universitätsmedizin Berlin; Berlin 10117 Germany
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry; Charité-Universitätsmedizin Berlin; Berlin 10117 Germany
- Berlin Institute of Health (BIH); Berlin Germany
- Cluster of Excellence NeuroCure and DZNE; Berlin 10117 Germany
| |
Collapse
|
185
|
Chiurchiù V, Leuti A, Cencioni MT, Albanese M, De Bardi M, Bisogno T, Centonze D, Battistini L, Maccarrone M. Modulation of monocytes by bioactive lipid anandamide in multiple sclerosis involves distinct Toll-like receptors. Pharmacol Res 2016; 113:313-319. [DOI: 10.1016/j.phrs.2016.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 01/01/2023]
|
186
|
Oosterhof N, Holtman IR, Kuil LE, van der Linde HC, Boddeke EWGM, Eggen BJL, van Ham TJ. Identification of a conserved and acute neurodegeneration-specific microglial transcriptome in the zebrafish. Glia 2016; 65:138-149. [PMID: 27757989 PMCID: PMC5215681 DOI: 10.1002/glia.23083] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/21/2016] [Accepted: 09/28/2016] [Indexed: 12/29/2022]
Abstract
Microglia are brain resident macrophages important for brain development, connectivity, homeostasis and disease. However, it is still largely unclear how microglia functions and their identity are regulated at the molecular level. Although recent transcriptomic studies have identified genes specifically expressed in microglia, the function of most of these genes in microglia is still unknown. Here, we performed RNA sequencing on microglia acutely isolated from healthy and neurodegenerative zebrafish brains. We found that a large fraction of the mouse microglial signature is conserved in the zebrafish, corroborating the use of zebrafish to help understand microglial genetics in mammals in addition to studying basic microglia biology. Second, our transcriptome analysis of microglia following neuronal ablation suggested primarily a proliferative response of microglia, which we confirmed by immunohistochemistry and in vivo imaging. Together with the recent improvements in genome editing technology in zebrafish, these data offer opportunities to facilitate functional genetic research on microglia in vivo in the healthy as well as in the diseased brain. GLIA 2016;65:138–149
Collapse
Affiliation(s)
- Nynke Oosterhof
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Wytemaweg 80, CN, 3015, The Netherlands
| | - Inge R Holtman
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 971 3, AV, Groningen, The Netherlands
| | - Laura E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Wytemaweg 80, CN, 3015, The Netherlands
| | - Herma C van der Linde
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Wytemaweg 80, CN, 3015, The Netherlands
| | - Erik W G M Boddeke
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 971 3, AV, Groningen, The Netherlands
| | - Bart J L Eggen
- Department of Neuroscience, Section Medical Physiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 971 3, AV, Groningen, The Netherlands
| | - Tjakko J van Ham
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Wytemaweg 80, CN, 3015, The Netherlands
| |
Collapse
|
187
|
Renner K, Hellerbrand S, Hermann F, Riedhammer C, Talke Y, Schiechl G, Rodriguez Gomez M, Kutzi S, Halbritter D, Goebel N, Brühl H, Weissert R, Mack M. IL-3 promotes the development of experimental autoimmune encephalitis. JCI Insight 2016; 1:e87157. [PMID: 27734026 DOI: 10.1172/jci.insight.87157] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Little is known about the role of IL-3 in multiple sclerosis (MS) in humans and in experimental autoimmune encephalomyelitis (EAE). Using myelin oligodendrocyte glycoprotein (MOG) peptide-induced EAE, we show that CD4+ T cells are the main source of IL-3 and that cerebral IL-3 expression correlates with the influx of T cells into the brain. Blockade of IL-3 with monoclonal antibodies, analysis of IL-3 deficient mice, and adoptive transfer of leukocytes demonstrate that IL-3 plays an important role for development of clinical symptoms of EAE, for migration of leukocytes into the brain, and for cerebral expression of adhesion molecules and chemokines. In contrast, injection of recombinant IL-3 exacerbates EAE symptoms and cerebral inflammation. In patients with relapsing-remitting MS (RRMS), IL-3 expression by T cells is markedly upregulated during episodes of relapse. Our data indicate that IL-3 plays an important role in EAE and may represent a new target for treatment of MS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hilke Brühl
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Robert Weissert
- Department of Neurology.,Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Matthias Mack
- Department of Internal Medicine II - Nephrology.,Regensburg Center for Interventional Immunology, Regensburg, Germany
| |
Collapse
|
188
|
Kamphuis W, Kooijman L, Schetters S, Orre M, Hol EM. Transcriptional profiling of CD11c-positive microglia accumulating around amyloid plaques in a mouse model for Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1847-60. [DOI: 10.1016/j.bbadis.2016.07.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/01/2016] [Accepted: 07/13/2016] [Indexed: 12/25/2022]
|
189
|
Infiltrating monocytes promote brain inflammation and exacerbate neuronal damage after status epilepticus. Proc Natl Acad Sci U S A 2016; 113:E5665-74. [PMID: 27601660 DOI: 10.1073/pnas.1604263113] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The generalized seizures of status epilepticus (SE) trigger a series of molecular and cellular events that produce cognitive deficits and can culminate in the development of epilepsy. Known early events include opening of the blood-brain barrier (BBB) and astrocytosis accompanied by activation of brain microglia. Whereas circulating monocytes do not infiltrate the healthy CNS, monocytes can enter the brain in response to injury and contribute to the immune response. We examined the cellular components of innate immune inflammation in the days following SE by discriminating microglia vs. brain-infiltrating monocytes. Chemokine receptor 2 (CCR2(+)) monocytes invade the hippocampus between 1 and 3 d after SE. In contrast, only an occasional CD3(+) T lymphocyte was encountered 3 d after SE. The initial cellular sources of the chemokine CCL2, a ligand for CCR2, included perivascular macrophages and microglia. The induction of the proinflammatory cytokine IL-1β was greater in FACS-isolated microglia than in brain-invading monocytes. However, Ccr2 knockout mice displayed greatly reduced monocyte recruitment into brain and reduced levels of the proinflammatory cytokine IL-1β in hippocampus after SE, which was explained by higher expression of the cytokine in circulating and brain monocytes in wild-type mice. Importantly, preventing monocyte recruitment accelerated weight regain, reduced BBB degradation, and attenuated neuronal damage. Our findings identify brain-infiltrating monocytes as a myeloid-cell subclass that contributes to neuroinflammation and morbidity after SE. Inhibiting brain invasion of CCR2(+) monocytes could represent a viable method for alleviating the deleterious consequences of SE.
Collapse
|
190
|
Andreasson KI, Bachstetter AD, Colonna M, Ginhoux F, Holmes C, Lamb B, Landreth G, Lee DC, Low D, Lynch MA, Monsonego A, O’Banion MK, Pekny M, Puschmann T, Russek-Blum N, Sandusky LA, Selenica MLB, Takata K, Teeling J, Town T, Van Eldik LJ, Russek-Blum N, Monsonego A, Low D, Takata K, Ginhoux F, Town T, O’Banion MK, Lamb B, Colonna M, Landreth G, Andreasson KI, Sandusky LA, Selenica MLB, Lee DC, Holmes C, Teeling J, Lynch MA, Van Eldik LJ, Bachstetter AD, Pekny M, Puschmann T. Targeting innate immunity for neurodegenerative disorders of the central nervous system. J Neurochem 2016; 138:653-93. [PMID: 27248001 PMCID: PMC5433264 DOI: 10.1111/jnc.13667] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/01/2016] [Accepted: 04/30/2016] [Indexed: 12/21/2022]
Abstract
Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview of physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia and astrocyte cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article. Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer's disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview on physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Niva Russek-Blum
- The Dead Sea and Arava Science Center, Central Arava Branch, Yair Station, Hazeva, Israel
| | - Alon Monsonego
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, The Faculty of Health Sciences: The National Institute of Biotechnology in the Negev, and Zlotowski Center for Neuroscience, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kazuyuki Takata
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Terrence Town
- Departments of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089,
| | - M. Kerry O’Banion
- Departments of Neuroscience and Neurology, Del Monte Neuromedicine Institute, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642,
| | - Bruce Lamb
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH 44106
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gary Landreth
- Department of Neurosciences, Case Western Reserve University 44106
| | - Katrin I. Andreasson
- Department of Neurology and Neurological Sciences, Stanford Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leslie A. Sandusky
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Maj-Linda B. Selenica
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Daniel C. Lee
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Clive Holmes
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | - Jessica Teeling
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | | | | | | | - Milos Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| | - Till Puschmann
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
191
|
Hucke S, Herold M, Liebmann M, Freise N, Lindner M, Fleck AK, Zenker S, Thiebes S, Fernandez-Orth J, Buck D, Luessi F, Meuth SG, Zipp F, Hemmer B, Engel DR, Roth J, Kuhlmann T, Wiendl H, Klotz L. The farnesoid-X-receptor in myeloid cells controls CNS autoimmunity in an IL-10-dependent fashion. Acta Neuropathol 2016; 132:413-31. [PMID: 27383204 DOI: 10.1007/s00401-016-1593-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 06/09/2016] [Accepted: 06/26/2016] [Indexed: 12/24/2022]
Abstract
Innate immune responses by myeloid cells decisively contribute to perpetuation of central nervous system (CNS) autoimmunity and their pharmacologic modulation represents a promising strategy to prevent disease progression in Multiple Sclerosis (MS). Based on our observation that peripheral immune cells from relapsing-remitting and primary progressive MS patients exhibited strongly decreased levels of the bile acid receptor FXR (farnesoid-X-receptor, NR1H4), we evaluated its potential relevance as therapeutic target for control of established CNS autoimmunity. Pharmacological FXR activation promoted generation of anti-inflammatory macrophages characterized by arginase-1, increased IL-10 production, and suppression of T cell responses. In mice, FXR activation ameliorated CNS autoimmunity in an IL-10-dependent fashion and even suppressed advanced clinical disease upon therapeutic administration. In analogy to rodents, pharmacological FXR activation in human monocytes from healthy controls and MS patients induced an anti-inflammatory phenotype with suppressive properties including control of effector T cell proliferation. We therefore, propose an important role of FXR in control of T cell-mediated autoimmunity by promoting anti-inflammatory macrophage responses.
Collapse
Affiliation(s)
- Stephanie Hucke
- Department of Neurology, University of Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
| | - Martin Herold
- Department of Neurology, University of Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
| | - Marie Liebmann
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Nicole Freise
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Maren Lindner
- Department of Neurology, University of Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
| | - Ann-Katrin Fleck
- Department of Neurology, University of Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
| | - Stefanie Zenker
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Stephanie Thiebes
- Institute of Experimental Immunology and Imaging, University Duisburg-Essen, Essen, Germany
| | - Juncal Fernandez-Orth
- Department of Neurology, University of Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
| | - Dorothea Buck
- Department of Neurology, Technische Universität München, Munich, Germany
| | - Felix Luessi
- Department of Neurology, University of Mainz, Mainz, Germany
| | - Sven G Meuth
- Department of Neurology, University of Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
- Cluster of Excellence Cells in Motion, University of Muenster, Muenster, Germany
| | - Frauke Zipp
- Department of Neurology, University of Mainz, Mainz, Germany
| | - Bernhard Hemmer
- Department of Neurology, Technische Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Daniel Robert Engel
- Institute of Experimental Immunology and Imaging, University Duisburg-Essen, Essen, Germany
| | - Johannes Roth
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University of Muenster, Muenster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
- Cluster of Excellence Cells in Motion, University of Muenster, Muenster, Germany
| | - Luisa Klotz
- Department of Neurology, University of Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany.
| |
Collapse
|
192
|
McKinney A, Benson J, Nascene D, Eisengart J, Salmela M, Loes D, Zhang L, Patel K, Raymond G, Miller W. Childhood Cerebral Adrenoleukodystrophy: MR Perfusion Measurements and Their Use in Predicting Clinical Outcome after Hematopoietic Stem Cell Transplantation. AJNR Am J Neuroradiol 2016; 37:1713-20. [PMID: 27079370 PMCID: PMC5018408 DOI: 10.3174/ajnr.a4773] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 02/08/2016] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND PURPOSE MR perfusion has shown abnormalities of affected WM in cerebral X-linked adrenoleukodystrophy, but serial data is needed to explore the import of such findings after hematopoietic stem cell transplantation. Our aim was to prospectively measure MR perfusion parameters in patients with cerebral adrenoleukodystrophy pre- and post-hematopoietic stem cell transplantation, and to correlate those measurements with clinical outcome. MATERIALS AND METHODS Ten patients with cerebral adrenoleukodystrophy prospectively underwent DSC-MR perfusion imaging at <45 days pre- (baseline), 30-60 days post-, and 1 year post-hematopoietic stem cell transplantation. MR perfusion measurements in the 10 patients and 8 controls were obtained from the parieto-occipital WM, splenium of the corpus callosum, leading enhancing edge, and normal-appearing frontal white matter. MR imaging severity scores and clinical neurologic function and neurocognitive scores were also obtained. MR perfusion values were analyzed in the patients with cerebral adrenoleukodystrophy at each time point and compared with those in controls. Correlations were calculated between the pre-hematopoietic stem cell transplantation MR perfusion values and 1-year clinical scores, with P value adjustment for multiple comparisons. RESULTS At baseline in patients with cerebral adrenoleukodystrophy, both relative CBV and relative CBF within the splenium of the corpus callosum and parieto-occipital WM significantly differed from those in controls (P = .005-.031) and remained so 1 year post-hematopoietic stem cell transplantation (P = .003-.005). Meanwhile, no MR perfusion parameter within the leading enhancing edge differed significantly from that in controls at baseline or at 1 year (P = .074-.999) or significantly changed by 1 year post-hematopoietic stem cell transplantation (P = .142-.887). Baseline Loes scores correlated with 1-year clinical neurologic function (r = 0.813, P < .0001), while splenium of the corpus callosum relative CBV also significantly correlated with 1-year neurologic function scale and the neurocognitive full-scale intelligence quotient and performance intelligence quotient scores (r = -0.730-0.815, P = .007-.038). CONCLUSIONS Leading enhancing edge measurements likely remain normal post-hematopoietic stem cell transplantation in cerebral adrenoleukodystrophy, suggesting local disease stabilization. Meanwhile, parieto-occipital WM and splenium of the corpus callosum relative CBV and relative CBF values worsened; this change signified irreversible injury. Baseline splenium of the corpus callosum relative CBV may predict clinical outcomes following hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- A.M. McKinney
- From the Departments of Radiology (A.M.M., J.B., D.R.N., M.B.S.)
| | - J. Benson
- From the Departments of Radiology (A.M.M., J.B., D.R.N., M.B.S.)
| | - D.R. Nascene
- From the Departments of Radiology (A.M.M., J.B., D.R.N., M.B.S.)
| | - J. Eisengart
- Pediatrics (J.E., G.R.), Division of Clinical Behavioral Neuroscience, University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota
| | - M.B. Salmela
- From the Departments of Radiology (A.M.M., J.B., D.R.N., M.B.S.)
| | - D.J. Loes
- Suburban Radiologic Consultants (D.J.L.), Minneapolis, Minnesota
| | - L. Zhang
- Clinical and Translational Science Institute (L.Z.), University of Minnesota, Minneapolis, Minnesota
| | - K. Patel
- Radiology Associates of the Fox Valley (K.P.), Neenah, Wisconsin
| | - G.V. Raymond
- Pediatrics (J.E., G.R.), Division of Clinical Behavioral Neuroscience, University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota
| | - W.P. Miller
- Department of Pediatrics (W.P.M.), Pediatric Blood and Marrow Transplantation Division, University of Minnesota Amplatz Children's Hospital, Minneapolis, Minnesota
| |
Collapse
|
193
|
Abstract
Discussions of multiple sclerosis (MS) pathophysiology tend to focus on T cells and B cells of the adaptive immune response. The innate immune system is less commonly considered in this context, although dendritic cells, monocytes, macrophages and microglia - collectively referred to as myeloid cells - have prominent roles in MS pathogenesis. These populations of myeloid cells function as antigen-presenting cells and effector cells in neuroinflammation. Furthermore, a vicious cycle of interactions between T cells and myeloid cells exacerbates pathology. Several disease-modifying therapies are now available to treat MS, and insights into their mechanisms of action have largely focused on the adaptive immune system, but these therapies also have important effects on myeloid cells. In this Review, we discuss the evidence for the roles of myeloid cells in MS and the experimental autoimmune encephalomyelitis model of MS, and consider how interactions between myeloid cells and T cells and/or B cells promote MS pathology. Finally, we discuss the direct and indirect effects of existing MS medications on myeloid cells.
Collapse
Affiliation(s)
- Manoj K Mishra
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
194
|
Boros P, Ochando J, Zeher M. Myeloid derived suppressor cells and autoimmunity. Hum Immunol 2016; 77:631-636. [DOI: 10.1016/j.humimm.2016.05.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/26/2016] [Accepted: 05/26/2016] [Indexed: 12/13/2022]
|
195
|
Parsa R, Lund H, Tosevski I, Zhang XM, Malipiero U, Beckervordersandforth J, Merkler D, Prinz M, Gyllenberg A, James T, Warnecke A, Hillert J, Alfredsson L, Kockum I, Olsson T, Fontana A, Suter T, Harris RA. TGFβ regulates persistent neuroinflammation by controlling Th1 polarization and ROS production via monocyte-derived dendritic cells. Glia 2016; 64:1925-37. [PMID: 27479807 PMCID: PMC5053226 DOI: 10.1002/glia.23033] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/07/2016] [Indexed: 12/15/2022]
Abstract
Intracerebral levels of Transforming Growth Factor beta (TGFβ) rise rapidly during the onset of experimental autoimmune encephalomyelitis (EAE), a mouse model of Multiple Sclerosis (MS). We addressed the role of TGFβ responsiveness in EAE by targeting the TGFβ receptor in myeloid cells, determining that Tgfbr2 was specifically targeted in monocyte‐derived dendritic cells (moDCs) but not in CNS resident microglia by using bone‐marrow chimeric mice. TGFβ responsiveness in moDCs was necessary for the remission phase since LysMCreTgfbr2fl/fl mice developed a chronic form of EAE characterized by severe demyelination and extensive infiltration of activated moDCs in the CNS. Tgfbr2 deficiency resulted in increased moDC IL‐12 secretion that skewed T cells to produce IFN‐γ, which in turn enhanced the production of moDC‐derived reactive oxygen species that promote oxidative damage and demyelination. We identified SNPs in the human NOX2 (CYBB) gene that associated with the severity of MS, and significantly increased CYBB expression was recorded in PBMCs from both MS patients and from MS severity risk allele rs72619425‐A carrying individuals. We thus identify a novel myeloid cell‐T cell activation loop active in the CNS during chronic disease that could be therapeutically targeted. GLIA 2016;64:1925–1937
Collapse
Affiliation(s)
- Roham Parsa
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Harald Lund
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Ivana Tosevski
- Clinical Immunology, University Hospital of Zurich, Switzerland
| | - Xing-Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | | | - Jan Beckervordersandforth
- Institute of Neuropathology and BIOSS Centre for Biological Signaling Studies, University of Freiburg, Germany.,Department of Neuropathology, Georg-August-University Goettingen, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, and; Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Marco Prinz
- Department of Neuropathology, Georg-August-University Goettingen, Germany
| | - Alexandra Gyllenberg
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Tojo James
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Andreas Warnecke
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Jan Hillert
- Neurogenetics Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Lars Alfredsson
- Cardiovascular Epidemiology Unit, Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Adriano Fontana
- Clinical Immunology, University Hospital of Zurich, Switzerland.,Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Tobias Suter
- Clinical Immunology, University Hospital of Zurich, Switzerland.,Department of Neurology and Clinical Research Priority Program Multiple Sclerosis, University Hospital Zurich, Switzerland
| | - Robert A Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Centre for Molecular Medicine, Karolinska University Hospital, Solna, Sweden.
| |
Collapse
|
196
|
Lee MJ, Bing SJ, Choi J, Jang M, Lee G, Lee H, Chang BS, Jee Y, Lee SJ, Cho IH. IKKβ-mediated inflammatory myeloid cell activation exacerbates experimental autoimmune encephalomyelitis by potentiating Th1/Th17 cell activation and compromising blood brain barrier. Mol Neurodegener 2016; 11:54. [PMID: 27450563 PMCID: PMC4957872 DOI: 10.1186/s13024-016-0116-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 07/02/2016] [Indexed: 01/31/2023] Open
Abstract
Background The inflammatory myeloid cell activation is one of the hallmarks of experimental autoimmune encephalomyelitis (EAE), yet the in vivo role of the inflammatory myeloid cell activation in EAE has not been clearly resolved. It is well-known that IKK/NF-κB is a key signaling pathway that regulates inflammatory myeloid activation. Methods We investigated the in vivo role of inflammatory myeloid cell activation in myelin oligodendrocyte glycoprotein (MOG) peptides-induced EAE using myeloid cell type-specific ikkβ gene conditional knockout-mice (LysM-Cre/IkkβF/F). Results In our study, LysM-Cre/IkkβF/F mice had alleviated clinical signs of EAE corresponding to the decreased spinal demyelination, microglial activation, and immune cell infiltration in the spinal cord, compared to the wild-type mice (WT, IkkβF/F). Myeloid ikkβ gene deletion significantly reduced the percentage of CD4+/IFN-γ+ (Th1) and CD4+/IL-17+ (Th17) cells but increased the percentages of CD4+/CD25+/Foxp3+ (Treg) cells in the spinal cord and lymph nodes, corresponding to the altered mRNA expression of IFN-γ, IL-17, IL-23, and Foxp3 in the spinal cords of LysM-Cre/IkkβF/F EAE mice. Also, the beneficial effect of myeloid IKKβ deletion in EAE corresponded to the decreased permeability of the blood brain barrier (BBB). Conclusions Our findings strongly suggest that IKK/NF-kB-induced myeloid cell activation exacerbates EAE by activating Th1 and Th17 responses and compromising the BBB. The development of NF-κB inhibitory agents with high efficacy through specific targeting of IKKβ in myeloid cells might be of therapeutic potential in MS and other autoimmune disorders. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0116-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Jung Lee
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.,Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - So Jin Bing
- Department of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jonghee Choi
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.,Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Minhee Jang
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Gihyun Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyunkyoung Lee
- Department of Neuroscience and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung Soo Chang
- Department of Cosmetology, Hanseo University, Seosan, 31962, Republic of Korea
| | - Youngheun Jee
- Department of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Sung Joong Lee
- Department of Neuroscience and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea. .,Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea. .,Institute of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
197
|
Davies CL, Miron VE. Distinct origins, gene expression and function of microglia and monocyte-derived macrophages in CNS myelin injury and regeneration. Clin Immunol 2016; 189:57-62. [PMID: 27377535 DOI: 10.1016/j.clim.2016.06.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/27/2016] [Accepted: 06/30/2016] [Indexed: 01/07/2023]
Abstract
Central nervous system (CNS) injury incurs a rapid innate immune response, including that from macrophages derived from endogenous microglia and circulating monocytes infiltrating the lesion site. One example of such injury is the demyelination observed in the autoimmune disease multiple sclerosis (MS), where macrophages are implicated in both myelin injury and regeneration. Although initially microglia and monocyte-derived macrophages were considered to have identical origins, gene expression, and function, recent advances have revealed important distinctions in all three categories and have caused a paradigm shift in view of their unique identity and roles. This has important consequences for understanding their individual contribution to neurological function and therapeutic targeting of these populations in diseases like MS. Here, we address the differences between CNS endogenous and exogenously-derived macrophages with a particular focus on myelin damage and regeneration.
Collapse
Affiliation(s)
- Claire L Davies
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | - Veronique E Miron
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| |
Collapse
|
198
|
Infiltrating T lymphocytes reduce myeloid phagocytosis activity in synucleinopathy model. J Neuroinflammation 2016; 13:174. [PMID: 27364890 PMCID: PMC4929755 DOI: 10.1186/s12974-016-0632-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/20/2016] [Indexed: 01/25/2023] Open
Abstract
Background Synucleinopathies comprise a group of neurodegenerative diseases associated with abnormal accumulation of α-synuclein. One of the key factors that contribute to the progression of synucleinopathies is neuroinflammation. However, the role of lymphocytes in synucleinopathies like Parkinson’s disease (PD) remains largely unclear. Methods To investigate how lymphocytes impact synucleinopathies, human wild-type α-synuclein (WTS) transgenic mice were crossed with mice lacking mature lymphocytes (Rag2−/−). In this in vivo model, we quantified α-synuclein aggregation in the substantia nigra (SN) and striatum and determined the numbers of innate and adaptive immune cells in the central nervous system (CNS). The activation state of resident and infiltrated CNS myeloid cells (M1 vs. M2) was further classified by gene and protein expression analyses. The impact of T and B lymphocytes on the phagocytic activity of microglia in the presence of α-synuclein aggregates was addressed in BV2 microglia in vitro. Results Compared to WTS+ Rag2+/+ mice, where T but not B lymphocytes infiltrated the CNS, decreased amounts of α-synuclein aggregates were found in WTS+ Rag2−/− mice devoid of mature lymphocytes. The presence of T lymphocytes did not alter the number of Iba1+ microglia but increased the frequency of the CD11b+ CD45hi population in the CNS, indicative of an increased number of infiltrated macrophages. Moreover, the M1 phenotype was more prominent in WTS+ Rag2+/+ mice, whereas the M2 activation state was dominating in the absence of lymphocytes in WTS+ Rag2−/− mice. In vitro, in the presence of T but not B lymphocytes, significantly less α-synuclein was phagocytosed by BV2 microglia, further supporting the prevalence of the M1 phenotype in the presence of T lymphocytes. Conclusions Peripheral T lymphocytes strongly contribute to increased α-synuclein pathology via modulation of CNS myeloid cell function. In the presence of T lymphocytes, microglia phagocytosis of aggregated α-synuclein is reduced, which increases the severity of synucleinopathy. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0632-5) contains supplementary material, which is available to authorized users.
Collapse
|
199
|
Mangalam AK, Rattan R, Suhail H, Singh J, Hoda MN, Deshpande M, Fulzele S, Denic A, Shridhar V, Kumar A, Viollet B, Rodriguez M, Giri S. AMP-Activated Protein Kinase Suppresses Autoimmune Central Nervous System Disease by Regulating M1-Type Macrophage–Th17 Axis. THE JOURNAL OF IMMUNOLOGY 2016; 197:747-60. [DOI: 10.4049/jimmunol.1501549] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 05/31/2016] [Indexed: 01/22/2023]
|
200
|
Friend or Foe? Resident Microglia vs Bone Marrow-Derived Microglia and Their Roles in the Retinal Degeneration. Mol Neurobiol 2016; 54:4094-4112. [PMID: 27318678 DOI: 10.1007/s12035-016-9960-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/06/2016] [Indexed: 01/10/2023]
Abstract
Microglia are immune cells in the central nervous system (CNS) that originate from the yolk sac in an embryo. The renewal of the microglia pool in the adult eye consists of two components. In addition to the self-proliferation of resident cells, microglia in the CNS also derive from the bone marrow (BM). BM-derived cells pass through the blood-brain barrier (BBB) or blood-retina barrier (BRB) and differentiate into microglia under specific conditions which involves a complex mechanism. Recent studies have widely investigated the role of resident microglia and BM-derived microglia in the retinal degenerative disease. Both two cell types play dual roles and share many similar functions. However, resident microglia tend to polarize to the M1 phenotype which is pro-inflammatory and neurotoxic, whereas BM-derived microglia mainly polarize to the neuroprotective M2 phenotype in retinal degeneration. The molecular mechanism that underlines the invasion of peripheral cells has led to extensive discussions. In addition to the BBB and BRB disruption, many signaling pathways are involved in this process. Based on these studies, we discuss the roles of these two types of microglia in retinal degeneration disease and the potential clinical application of BM-derived microglia, which may benefit future therapies.
Collapse
|