151
|
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
|
152
|
Lovelace MD, Varney B, Sundaram G, Franco NF, Ng ML, Pai S, Lim CK, Guillemin GJ, Brew BJ. Current Evidence for a Role of the Kynurenine Pathway of Tryptophan Metabolism in Multiple Sclerosis. Front Immunol 2016; 7:246. [PMID: 27540379 PMCID: PMC4972824 DOI: 10.3389/fimmu.2016.00246] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022] Open
Abstract
The kynurenine pathway (KP) is the major metabolic pathway of the essential amino acid tryptophan (TRP). Stimulation by inflammatory molecules, such as interferon-γ (IFN-γ), is the trigger for induction of the KP, driving a complex cascade of production of both neuroprotective and neurotoxic metabolites, and in turn, regulation of the immune response and responses of brain cells to the KP metabolites. Consequently, substantial evidence has accumulated over the past couple of decades that dysregulation of the KP and the production of neurotoxic metabolites are associated with many neuroinflammatory and neurodegenerative diseases, including Parkinson’s disease, AIDS-related dementia, motor neurone disease, schizophrenia, Huntington’s disease, and brain cancers. In the past decade, evidence of the link between the KP and multiple sclerosis (MS) has rapidly grown and has implicated the KP in MS pathogenesis. KP enzymes, indoleamine 2,3-dioxygenase (IDO-1) and tryptophan dioxygenase (highest expression in hepatic cells), are the principal enzymes triggering activation of the KP to produce kynurenine from TRP. This is in preference to other routes such as serotonin and melatonin production. In neurological disease, degradation of the blood–brain barrier, even if transient, allows the entry of blood monocytes into the brain parenchyma. Similar to microglia and macrophages, these cells are highly responsive to IFN-γ, which upregulates the expression of enzymes, including IDO-1, producing neurotoxic KP metabolites such as quinolinic acid. These metabolites circulate systemically or are released locally in the brain and can contribute to the excitotoxic death of oligodendrocytes and neurons in neurological disease principally by virtue of their agonist activity at N-methyl-d-aspartic acid receptors. The latest evidence is presented and discussed. The enzymes that control the checkpoints in the KP represent an attractive therapeutic target, and consequently several KP inhibitors are currently in clinical trials for other neurological diseases, and hence may make suitable candidates for MS patients. Underpinning these drug discovery endeavors, in recent years, several advances have been made in how KP metabolites are assayed in various biological fluids, and tremendous advancements have been made in how specimens are imaged to determine disease progression and involvement of various cell types and molecules in MS.
Collapse
Affiliation(s)
- Michael D Lovelace
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Bianca Varney
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Gayathri Sundaram
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Nunzio F Franco
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Mei Li Ng
- Faculty of Medicine, Sydney Medical School, University of Sydney , Sydney, NSW , Australia
| | - Saparna Pai
- Sydney Medical School, University of Sydney , Sydney, NSW , Australia
| | - Chai K Lim
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University , Sydney, NSW , Australia
| | - Gilles J Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University , Sydney, NSW , Australia
| | - Bruce J Brew
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia; Department of Neurology, St Vincent's Hospital, Sydney, NSW, Australia
| |
Collapse
|
153
|
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: 5.7] [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]
|
154
|
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: 2.3] [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
|
155
|
Chuang TY, Guo Y, Seki SM, Rosen AM, Johanson DM, Mandell JW, Lucchinetti CF, Gaultier A. LRP1 expression in microglia is protective during CNS autoimmunity. Acta Neuropathol Commun 2016; 4:68. [PMID: 27400748 PMCID: PMC4940960 DOI: 10.1186/s40478-016-0343-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 06/28/2016] [Indexed: 01/09/2023] Open
Abstract
Multiple sclerosis is a devastating neurological disorder characterized by the autoimmune destruction of the central nervous system myelin. While T cells are known orchestrators of the immune response leading to MS pathology, the precise contribution of CNS resident and peripheral infiltrating myeloid cells is less well described. Here, we explore the myeloid cell function of Low-density lipoprotein receptor-related protein-1 (LRP1), a scavenger receptor involved in myelin clearance and the inflammatory response, in the context of Multiple sclerosis. Supporting its central role in Multiple sclerosis pathology, we find that LRP1 expression is increased in Multiple sclerosis lesions in comparison to the surrounding healthy tissue. Using two genetic mouse models, we show that deletion of LRP1 in microglia, but not in peripheral macrophages, negatively impacts the progression of experimental autoimmune encephalomyelitis, an animal model of Multiple sclerosis. We further show that the increased disease severity in experimental autoimmune encephalomyelitis is not due to haplodeficiency of the Cx3cr1 locus. At the cellular level, microglia lacking LRP1 adopt a pro-inflammatory phenotype characterized by amoeboid morphology and increased production of the inflammatory mediator TNF-α. We also show that LRP1 functions as a robust inhibitor of NF-kB activation in myeloid cells via a MyD88 dependent pathway, potentially explaining the increase in disease severity observed in mice lacking LRP1 expression in microglia. Taken together, our data suggest that the function of LRP1 in microglia is to keep these cells in an anti-inflammatory and neuroprotective status during inflammatory insult, including experimental autoimmune encephalomyelitis and potentially in Multiple sclerosis.
Collapse
|
156
|
Shiomi A, Usui T, Mimori T. GM-CSF as a therapeutic target in autoimmune diseases. Inflamm Regen 2016; 36:8. [PMID: 29259681 PMCID: PMC5725926 DOI: 10.1186/s41232-016-0014-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/10/2016] [Indexed: 12/23/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been known as a hematopoietic growth factor and immune modulator. Recent studies revealed that GM-CSF also had pro-inflammatory functions and contributed to the pathogenicity of Th17 cells in the development of Th17-mediated autoimmune diseases. GM-CSF inhibition in some animal models of autoimmune diseases showed significant beneficial effects. Therefore, several agents targeting GM-CSF are being developed and are expected to be a useful strategy for the treatment of autoimmune diseases. Particularly, in clinical trials for rheumatoid arthritis (RA) patients, GM-CSF inhibition showed rapid and significant efficacy with no serious side effects. This article summarizes recent findings of GM-CSF and information of clinical trials targeting GM-CSF in autoimmune diseases.
Collapse
Affiliation(s)
- Aoi Shiomi
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54-Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Takashi Usui
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54-Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan
| | - Tsuneyo Mimori
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54-Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507 Japan
| |
Collapse
|
157
|
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: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 05/31/2016] [Indexed: 01/22/2023]
|
158
|
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: 3.4] [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
|
159
|
Foks AC, Van Puijvelde GHM, Wolbert J, Kröner MJ, Frodermann V, Van Der Heijden T, Van Santbrink PJ, Boon L, Bot I, Kuiper J. CD11b+Gr-1+ myeloid-derived suppressor cells reduce atherosclerotic lesion development in LDLr deficient mice. Cardiovasc Res 2016; 111:252-61. [PMID: 27234908 DOI: 10.1093/cvr/cvw114] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 05/16/2016] [Indexed: 01/25/2023] Open
Abstract
AIMS Myeloid-derived suppressor cells (MDSCs) form a heterogeneous population of cells composed of early myeloid progenitor cells and immature myeloid cells, which strongly suppress pro-inflammatory immune cells in inflammatory diseases. Currently, it is unknown whether MDSCs contribute to atherosclerosis, a chronic inflammatory disease in which accumulation of lipoproteins in the arterial wall activates the immune system causing abnormal vascular remodelling and vessel occlusion. Here, we investigated whether and how MDSCs contribute to the development of atherosclerosis. METHODS AND RESULTS We show that MDSCs arise in the bone marrow of LDLr(-/-) mice during atherosclerosis and strongly suppress proliferation of T cells. Adoptive transfer of MDSCs into both female and male LDLr(-/-) mice fed a Western-type diet (WTD) ameliorates atherosclerosis with 35%. We observed a 54% reduction in adventitial T cells, and more specifically, MDSCs suppress Th1 and Th17 cells. In addition, treatment with MDSCs reduces circulating pro-atherogenic B2 cells. We found two subsets of MDSCs in the bone marrow of hypercholesterolemic mice, monocytic and granulocytic MDSCs (mo- and gr-MDSCs, respectively), of which the percentage of mo-MDSCs significantly increased during WTD feeding. Moreover, mo-MDSCs completely abolished splenocyte proliferation, whereas gr-MDSCs were unable to suppress proliferation. Mechanistically, we show that MDSCs from atherosclerotic mice suppress T cells in an IFN-γ- and nitric oxide-dependent manner, which is associated with the action of mo-MDSCs. CONCLUSION This study demonstrates that MDSCs develop during atherosclerosis and reduce atherosclerosis via suppression of pro-inflammatory immune responses.
Collapse
Affiliation(s)
- Amanda C Foks
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Gijs H M Van Puijvelde
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Jolien Wolbert
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Mara J Kröner
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Vanessa Frodermann
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Thomas Van Der Heijden
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Peter J Van Santbrink
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | | | - Ilze Bot
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, Leiden 2333 CC, The Netherlands
| |
Collapse
|
160
|
Sniping the scout: Targeting the key molecules in dendritic cell functions for treatment of autoimmune diseases. Pharmacol Res 2016; 107:27-41. [DOI: 10.1016/j.phrs.2016.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 02/23/2016] [Accepted: 02/23/2016] [Indexed: 02/07/2023]
|
161
|
Wu H, Zhen Y, Ma Z, Li H, Yu J, Xu ZG, Wang XY, Yi H, Yang YG. Arginase-1-dependent promotion of TH17 differentiation and disease progression by MDSCs in systemic lupus erythematosus. Sci Transl Med 2016; 8:331ra40. [PMID: 27009269 PMCID: PMC4895207 DOI: 10.1126/scitranslmed.aae0482] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/16/2016] [Indexed: 12/22/2022]
Abstract
Expansion of myeloid-derived suppressor cells (MDSCs) has been documented in some murine models and patients with autoimmune diseases, but the exact role of MDSCs in this process remains largely unknown. The current study investigates this question in patients with systemic lupus erythematosus (SLE). Patients with active SLE showed a significant increase in HLA-DR(-)CD11b(+)CD33(+)MDSCs, including both CD14(+)CD66b(-)monocytic and CD14(-)CD66b(+)granulocytic MDSCs, in the peripheral blood compared to healthy controls (HCs). The frequency of MDSCs was positively correlated with the levels of serum arginase-1 (Arg-1) activity, T helper 17 (TH17) responses, and disease severity in SLE patients. Consistently, in comparison with MDSCs from HCs, MDSCs from SLE patients exhibited significantly elevated Arg-1 production and increased potential to promote TH17 differentiation in vitro in an Arg-1-dependent manner. Moreover, in a humanized SLE model, MDSCs were essential for the induction of TH17 responses and the associated renal injuries, and the effect of MDSCs was Arg-1-dependent. Our data provide direct evidence demonstrating a pathogenic role for MDSCs in human SLE. This study also provides a molecular mechanism of the pathogenesis of SLE by demonstrating an Arg-1-dependent effect of MDSCs in the development of TH17 cell-associated autoimmunity, and suggests that targeting MDSCs or Arg-1 may offer potential therapeutic strategies for the treatment of SLE and other TH17 cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Hao Wu
- The First Hospital and Institute of Immunology, Jilin University, Changchun 130061, China
| | - Yu Zhen
- The First Hospital and Institute of Immunology, Jilin University, Changchun 130061, China
| | - Zhanchuan Ma
- The First Hospital and Institute of Immunology, Jilin University, Changchun 130061, China
| | - Huimin Li
- The First Hospital and Institute of Immunology, Jilin University, Changchun 130061, China
| | - Jinyu Yu
- The First Hospital and Institute of Immunology, Jilin University, Changchun 130061, China
| | - Zhong-Gao Xu
- The First Hospital and Institute of Immunology, Jilin University, Changchun 130061, China
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Huanfa Yi
- The First Hospital and Institute of Immunology, Jilin University, Changchun 130061, China.
| | - Yong-Guang Yang
- The First Hospital and Institute of Immunology, Jilin University, Changchun 130061, China. Department of Medicine, Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
162
|
Bravo B, Gallego MI, Flores AI, Bornstein R, Puente-Bedia A, Hernández J, de la Torre P, García-Zaragoza E, Perez-Tavarez R, Grande J, Ballester A, Ballester S. Restrained Th17 response and myeloid cell infiltration into the central nervous system by human decidua-derived mesenchymal stem cells during experimental autoimmune encephalomyelitis. Stem Cell Res Ther 2016; 7:43. [PMID: 26987803 PMCID: PMC4797118 DOI: 10.1186/s13287-016-0304-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multiple sclerosis is a widespread inflammatory demyelinating disease. Several immunomodulatory therapies are available, including interferon-β, glatiramer acetate, natalizumab, fingolimod, and mitoxantrone. Although useful to delay disease progression, they do not provide a definitive cure and are associated with some undesirable side-effects. Accordingly, the search for new therapeutic methods constitutes an active investigation field. The use of mesenchymal stem cells (MSCs) to modify the disease course is currently the subject of intense interest. Decidua-derived MSCs (DMSCs) are a cell population obtained from human placental extraembryonic membranes able to differentiate into the three germ layers. This study explores the therapeutic potential of DMSCs. METHODS We used the experimental autoimmune encephalomyelitis (EAE) animal model to evaluate the effect of DMSCs on clinical signs of the disease and on the presence of inflammatory infiltrates in the central nervous system. We also compared the inflammatory profile of spleen T cells from DMSC-treated mice with that of EAE control animals, and the influence of DMSCs on the in vitro definition of the Th17 phenotype. Furthermore, we analyzed the effects on the presence of some critical cell types in central nervous system infiltrates. RESULTS Preventive intraperitoneal injection of DMSCs resulted in a significant delay of external signs of EAE. In addition, treatment of animals already presenting with moderate symptoms resulted in mild EAE with reduced disease scores. Besides decreased inflammatory infiltration, diminished percentages of CD4(+)IL17(+), CD11b(+)Ly6G(+) and CD11b(+)Ly6C(+) cells were found in infiltrates of treated animals. Early immune response was mitigated, with spleen cells of DMSC-treated mice displaying low proliferative response to antigen, decreased production of interleukin (IL)-17, and increased production of the anti-inflammatory cytokines IL-4 and IL-10. Moreover, lower RORγT and higher GATA-3 expression levels were detected in DMSC-treated mice. DMSCs also showed a detrimental influence on the in vitro definition of the Th17 phenotype. CONCLUSIONS DMSCs modulated the clinical course of EAE, modified the frequency and cell composition of the central nervous system infiltrates during the disease, and mediated an impairment of Th17 phenotype establishment in favor of the Th2 subtype. These results suggest that DMSCs might provide a new cell-based therapy for the control of multiple sclerosis.
Collapse
Affiliation(s)
- Beatriz Bravo
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Marta I. Gallego
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Mammary Gland Pathology, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Ana I. Flores
- Grupo de Medicina Regenerativa, Instituto de Investigación Hospital 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
| | - Rafael Bornstein
- Hospital Central de Cruz Roja, Servicio de Hematología y Hemoterapia, Avenida de Reina Victoria 24, 28003 Madrid, Spain
| | - Alba Puente-Bedia
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Javier Hernández
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Paz de la Torre
- Grupo de Medicina Regenerativa, Instituto de Investigación Hospital 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
| | - Elena García-Zaragoza
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Mammary Gland Pathology, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Raquel Perez-Tavarez
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Histology Core Unit, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Jesús Grande
- Grupo de Medicina Regenerativa, Instituto de Investigación Hospital 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
| | - Alicia Ballester
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Sara Ballester
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| |
Collapse
|
163
|
Gao Q, Zhang Y, Han C, Hu X, Zhang H, Xu X, Tian J, Liu Y, Ding Y, Liu J, Wang C, Guo Z, Yang Y, Cao X. Blockade of CD47 ameliorates autoimmune inflammation in CNS by suppressing IL-1-triggered infiltration of pathogenic Th17 cells. J Autoimmun 2016; 69:74-85. [PMID: 26994903 DOI: 10.1016/j.jaut.2016.03.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/04/2016] [Accepted: 03/06/2016] [Indexed: 12/11/2022]
Abstract
The migration of Th17 cells into central nervous system (CNS) tissue is the key pathogenic step in experimental autoimmune encephalomyelitis (EAE) model. However, the mechanism underlying the pathogenic Th17 cell migration remains elusive. Here we report that blockade of CD47 with CD47-Fc fusion protein is effective in preventing and curing EAE by impairing infiltration of Th17 cells into CNS. However, CD47 deficiency does not directly impair the migration of Th17 cells. Mechanistic studies showed that CD47 deficiency inhibited degradation of inducible nitric oxide synthase (iNOS) in proteasome of macrophages by Src activation and led to the increased nitric oxide (NO) production. Then NO suppressed inflammasome activation-induced IL-1β production. This lower IL-1β reduces the expression of IL-1R1 and migration-related chemokine receptors on CD47(-/-) Th17 cells, inhibiting the ability of Th17 cells to infiltrate into the CNS of CD47(-/-) mice and therefore suppressing EAE development. In vivo administration of exogenous IL-1β indeed promoted the infiltration CD47(-/-) Th17 cells into CNS and antagonized the protective role of CD47 deficiency in EAE pathogenesis. Our results demonstrate a potential preventive and therapeutic application of CD47 blockade in controlling EAE development.
Collapse
Affiliation(s)
- Qiangguo Gao
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China; Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China.
| | - Yi Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Chaofeng Han
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Xiang Hu
- National Key Laboratory of Medical Molecular Biology & Department of Immunology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Hua Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Xiongfei Xu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Jun Tian
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yiqi Liu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuanyuan Ding
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Juan Liu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Chunmei Wang
- National Key Laboratory of Medical Molecular Biology & Department of Immunology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Zhenhong Guo
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Yongguang Yang
- First Hospital of Jilin University, Changchun, 130012, China
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China; National Key Laboratory of Medical Molecular Biology & Department of Immunology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China; Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
164
|
|
165
|
Chemical dampening of Ly6C(hi) monocytes in the periphery produces anti-depressant effects in mice. Sci Rep 2016; 6:19406. [PMID: 26783261 PMCID: PMC4725984 DOI: 10.1038/srep19406] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 12/14/2015] [Indexed: 01/09/2023] Open
Abstract
The involvement of systemic immunity in depression pathogenesis promises a periphery-targeting paradigm in novel anti-depressant discovery. However, relatively little is known about druggable targets in the periphery for mental and behavioral control. Here we report that targeting Ly6Chi monocytes in blood can serve as a strategy for anti-depressant purpose. A natural compound, ginsenoside Rg1 (Rg1), was firstly validated as a periphery-restricted chemical probe. Rg1 selectively suppressed Ly6Chi monocytes recruitment to the inflamed mice brain. The proinflammatory potential of Ly6Chi monocytes to activate astrocytes was abrogated by Rg1, which led to a blunted feedback release of CCL2 to recruit the peripheral monocytes. In vitro study demonstrated that Rg1 pretreatment on activated THP-1 monocytes retarded their ability to trigger CCL2 secretion from co-cultured U251 MG astrocytes. CCL2-triggered p38/MAPK and PI3K/Akt activation were involved in the action of Rg1. Importantly, in mice models, we found that dampening Ly6Chi monocytes at the periphery ameliorated depression-like behavior induced by neuroinflammation or chronic social defeat stress. Together, our work unravels that blood Ly6Chi monocytes may serve as the target to enable remote intervention on the depressed brain, and identifies Rg1 as a lead compound for designing drugs targeting peripheral CCL2 signals.
Collapse
|
166
|
Ifergan I, Chen S, Zhang B, Miller SD. Cutting Edge: MicroRNA-223 Regulates Myeloid Dendritic Cell-Driven Th17 Responses in Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2016; 196:1455-1459. [PMID: 26783338 DOI: 10.4049/jimmunol.1501965] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/22/2015] [Indexed: 11/19/2022]
Abstract
Myeloid cells play a crucial role in the induction and sustained inflammation in neuroinflammatory disorders, such as multiple sclerosis. miR-223, a myeloid cell-specific microRNA, is one of the most upregulated microRNAs in multiple sclerosis patients. We demonstrate that miR-223-knockout mice display significantly reduced active and adoptive-transfer experimental autoimmune encephalomyelitis that is characterized by reduced numbers of myeloid dendritic cells (mDCs) and Th17 cells in the CNS. Knockout mDCs have increased PD-L1 and decreased IL-1β, IL-6, and IL-23 expression, as well as a reduced capacity to drive Th17, but not Th1, cell differentiation. Thus, miR-223 controls mDC-induced activation of pathologic Th17 responses during autoimmune inflammation.
Collapse
Affiliation(s)
- Igal Ifergan
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611.,Department of Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Siqi Chen
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611.,Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Bin Zhang
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611.,Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611.,Department of Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
167
|
Marro BS, Grist JJ, Lane TE. Inducible Expression of CXCL1 within the Central Nervous System Amplifies Viral-Induced Demyelination. THE JOURNAL OF IMMUNOLOGY 2016; 196:1855-64. [PMID: 26773148 DOI: 10.4049/jimmunol.1501802] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/16/2015] [Indexed: 12/11/2022]
Abstract
The functional role of the ELR(+) chemokine CXCL1 in host defense and disease following infection of the CNS with the neurotropic JHM strain of mouse hepatitis virus (JHMV) was examined. Mice in which expression of CXCL1 is under the control of a tetracycline-inducible promoter active within glial fibrillary acidic protein-positive cells were generated and this allowed for selectively increasing CNS expression of CXCL1 in response to JHMV infection and evaluating the effects on neuroinflammation, control of viral replication, and demyelination. Inducible expression of CNS-derived CXCL1 resulted in increased levels of CXCL1 protein within the serum, brain, and spinal cord that correlated with increased frequency of Ly6G(+)CD11b(+) neutrophils present within the CNS. Elevated levels of CXCL1 did not influence the generation of virus-specific T cells, and there was no difference in control of JHMV replication compared with control mice, indicating that T cell infiltration into the CNS is CXCL1-independent. Sustained CXCL1 expression within the CNS resulted in increased mortality that correlated with elevated neutrophil infiltration, diminished numbers of mature oligodendrocytes, and an increase in the severity of demyelination. Neutrophil ablation in CXCL1-transgenic mice reduced the severity of demyelination in mice, arguing for a role for these cells in white matter damage. Collectively, these findings illustrate that sustained CXCL1 expression amplifies the severity of white matter damage and that neutrophils can contribute to this process in a model of viral-induced neurologic disease.
Collapse
Affiliation(s)
- Brett S Marro
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine CA 92697; and
| | - Jonathan J Grist
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Thomas E Lane
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112
| |
Collapse
|
168
|
Puchta A, Naidoo A, Verschoor CP, Loukov D, Thevaranjan N, Mandur TS, Nguyen PS, Jordana M, Loeb M, Xing Z, Kobzik L, Larché MJ, Bowdish DME. TNF Drives Monocyte Dysfunction with Age and Results in Impaired Anti-pneumococcal Immunity. PLoS Pathog 2016; 12:e1005368. [PMID: 26766566 PMCID: PMC4713203 DOI: 10.1371/journal.ppat.1005368] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 12/06/2015] [Indexed: 12/24/2022] Open
Abstract
Monocyte phenotype and output changes with age, but why this occurs and how it impacts anti-bacterial immunity are not clear. We found that, in both humans and mice, circulating monocyte phenotype and function was altered with age due to increasing levels of TNF in the circulation that occur as part of the aging process. Ly6C+ monocytes from old (18–22 mo) mice and CD14+CD16+ intermediate/inflammatory monocytes from older adults also contributed to this “age-associated inflammation” as they produced more of the inflammatory cytokines IL6 and TNF in the steady state and when stimulated with bacterial products. Using an aged mouse model of pneumococcal colonization we found that chronic exposure to TNF with age altered the maturity of circulating monocytes, as measured by F4/80 expression, and this decrease in monocyte maturation was directly linked to susceptibility to infection. Ly6C+ monocytes from old mice had higher levels of CCR2 expression, which promoted premature egress from the bone marrow when challenged with Streptococcus pneumoniae. Although Ly6C+ monocyte recruitment and TNF levels in the blood and nasopharnyx were higher in old mice during S. pneumoniae colonization, bacterial clearance was impaired. Counterintuitively, elevated TNF and excessive monocyte recruitment in old mice contributed to impaired anti-pneumococcal immunity since bacterial clearance was improved upon pharmacological reduction of TNF or Ly6C+ monocytes, which were the major producers of TNF. Thus, with age TNF impairs inflammatory monocyte development, function and promotes premature egress, which contribute to systemic inflammation and is ultimately detrimental to anti-pneumococcal immunity. As we age, levels of inflammatory cytokines in the blood and tissues increase. Although this appears to be an inevitable part of aging, it ultimately contributes to declining health. Epidemiological studies indicate that older adults with higher than age-average levels of inflammatory cytokines are at increased risk of acquiring, becoming hospitalized with and dying of Streptococcus pneumoniae pneumonia but how age-associated inflammation increased susceptibility to was not entirely clear. We demonstrate that the increase in the inflammatory cytokine TNF that occurs with age cause monocytes to leave the bone marrow prematurely and these immature monocytes produce more inflammatory cytokines when stimulated with bacterial products, thus further increasing levels of inflammatory cytokines in the blood. Furthermore, although old mice have higher levels of these inflammatory monocytes arriving at the site of S. pneumoniae, they are not able to clear the bacteria. By pharmacologically or genetically removing the inflammatory cytokine TNF or reducing the number of inflammatory monocytes we were able to restore antibacterial immunity in aged mice. Thus we demonstrate that monocytes are both influenced by and contributors to age-associated inflammation and that chronic exposure to age-associated inflammation increases susceptibility to S. pneumoniae due to altering monocyte maturity and function.
Collapse
Affiliation(s)
- Alicja Puchta
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
| | - Avee Naidoo
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
| | - Chris P. Verschoor
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
| | - Dessi Loukov
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
| | - Netusha Thevaranjan
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
| | - Talveer S. Mandur
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Phuong-son Nguyen
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Manel Jordana
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Mark Loeb
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
- Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Canada
| | - Zhou Xing
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
| | - Lester Kobzik
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | | | - Dawn M. E. Bowdish
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
- * E-mail:
| |
Collapse
|
169
|
Therapeutic depletion of monocyte-derived cells protects from long-term axonal loss in experimental autoimmune encephalomyelitis. J Neuroimmunol 2016; 290:36-46. [DOI: 10.1016/j.jneuroim.2015.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 02/02/2023]
|
170
|
Ma H, Wan S, Xia CQ. Immunosuppressive CD11b+Ly6Chi monocytes in pristane-induced lupus mouse model. J Leukoc Biol 2015; 99:1121-9. [PMID: 26657791 DOI: 10.1189/jlb.3a0415-158r] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 11/16/2015] [Indexed: 12/24/2022] Open
Abstract
Myeloid-derived suppressor cells with immunosuppressive functions have been described to be associated with one of the mechanisms by which malignant tumors escape immune surveillance. However, little is known about the role of myeloid-derived suppressor cells in autoimmunity. In the current study, when we attempted to characterize the peritoneal cells in pristane-induced lupus model, as reported previously, we observed that there were markedly increased CD11b(+)Ly6C(hi) monocytes. Surprisingly, this type of monocytes was almost phenotypically identical to the reported monocytic myeloid-derived suppressor cells. Further analysis on how these CD11b(+)Ly6C(hi) cells affected T cell response showed that they strongly suppressed T cell proliferation in vitro in a manner dependent on cell-cell contact, NO, and PGE2. In addition, we found that CD11b(+)Ly6C(hi) monocytes inhibited Th1 differentiation but enhanced development of forkhead box p3(+)CD4(+) regulatory T cells. Consistent with the in vitro experimental results, the in vivo adoptive cell transfer study showed that infusion of pristane-treated syngeneic CD11b(+)Ly6C(hi) monocytes significantly suppressed the production of anti-keyhole limpet hemocyanin antibodies induced by keyhole limpet hemocyanin immunization. In addition, we found that CD11b(+)Ly6C(hi) monocytes were also increased significantly in spleen and peripheral blood and showed immunosuppressive characteristics similar to their peritoneal counterparts. Our findings indicate that CD11b(+)Ly6C(hi) monocytes in a pristane-induced lupus mouse model are monocytic myeloid-derived suppressor cells instead of inflammatory monocytes, as demonstrated previously. To our knowledge, this is the first to describe myeloid-derived suppressor cells in a pristane-induced lupus mouse model, which may lead to a better understanding of the role of CD11b(+)Ly6C(hi) monocytes in this specific pristane-induced lupus model.
Collapse
Affiliation(s)
- Huijuan Ma
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China; and
| | - Suigui Wan
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China; and
| | - Chang-Qing Xia
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, China; and Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
171
|
Chow KV, Lew AM, Sutherland RM, Zhan Y. Monocyte-Derived Dendritic Cells Promote Th Polarization, whereas Conventional Dendritic Cells Promote Th Proliferation. THE JOURNAL OF IMMUNOLOGY 2015; 196:624-36. [PMID: 26663720 DOI: 10.4049/jimmunol.1501202] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 11/10/2015] [Indexed: 12/31/2022]
Abstract
Monocyte-derived dendritic cells (moDCs) dramatically increase in numbers upon infection and inflammation; accordingly, we found that this also occurs during allogeneic responses. Despite their prominence, how emergent moDCs and resident conventional DCs (cDCs) divide their labor as APCs remain undefined. Hence, we compared both direct and indirect presentation by murine moDCs versus cDCs. We found that, despite having equivalent MHC class II expression and in vitro survival, moDCs were 20-fold less efficient than cDCs at inducing CD4(+) T cell proliferation through both direct and indirect Ag presentation. Despite this, moDCs were more potent at inducing Th1 and Th17 differentiation (e.g., 8-fold higher IFN-γ and 2-fold higher IL-17A in T cell cocultures), whereas cDCs induced 10-fold higher IL-2 production. Intriguingly, moDCs potently reduced the ability of cDCs to stimulate T cell proliferation in vitro and in vivo, partially through NO production. We surmise that such division of labor between moDCs and cDCs has implications for their respective roles in the immune response.
Collapse
Affiliation(s)
- Kevin V Chow
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia; Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; and
| | - Andrew M Lew
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia; Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Robyn M Sutherland
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yifan Zhan
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia;
| |
Collapse
|
172
|
Abstract
Granulocyte-macrophage colony stimulating factor (GM-CSF) is a growth factor first identified as an inducer of differentiation and proliferation of granulocytes and macrophages derived from haematopoietic progenitor cells. Later studies have shown that GM-CSF is involved in a wide range of biological processes in both innate and adaptive immunity, with its production being tightly linked to the response to danger signals. Given that the functions of GM-CSF span multiple tissues and biological processes, this cytokine has shown potential as a new and important therapeutic target in several autoimmune and inflammatory disorders - particularly in rheumatoid arthritis. Indeed, GM-CSF was one of the first cytokines detected in human synovial fluid from inflamed joints. Therapies that target GM-CSF or its receptor have been tested in preclinical studies with promising results, further supporting the potential of targeting the GM-CSF pathway. In this Review, we discuss our expanding view of the biology of GM-CSF, outline what has been learnt about GM-CSF from studies of animal models and human diseases, and summarize the results of early phase clinical trials evaluating GM-CSF antagonism in inflammatory disorders.
Collapse
|
173
|
Zheng R, Chen S, Chen S. Correlation between myeloid-derived suppressor cells and S100A8/A9 in tumor and autoimmune diseases. Int Immunopharmacol 2015; 29:919-925. [PMID: 26508452 DOI: 10.1016/j.intimp.2015.10.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 10/11/2015] [Accepted: 10/12/2015] [Indexed: 02/05/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that constitute an important component of immune regulatory system. Two calcium-binding proteins S100A8 and S100A9 act as important mediators in acute and chronic inflammation. In recent years, many researchers have found that MDSCs and S100A8/A9 operated with one another through a positive feedback loop to promote tumor development and metastasis. However, the correlation between MDSCs and S100A8/A9 in autoimmune diseases (AIDs) remains unknown. In this review, we discussed the co-operation of MDSCs and S100A8/A9 in tumor environment, and also, the role of these two components in AIDs.
Collapse
Affiliation(s)
- Ruoting Zheng
- Department of Endocrinology and Rheumatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China
| | - Shiyi Chen
- Department of Endocrinology and Rheumatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China
| | - Shenren Chen
- Department of Endocrinology and Rheumatology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China.
| |
Collapse
|
174
|
Almolda B, González B, Castellano B. Are Microglial Cells the Regulators of Lymphocyte Responses in the CNS? Front Cell Neurosci 2015; 9:440. [PMID: 26635525 PMCID: PMC4644801 DOI: 10.3389/fncel.2015.00440] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/23/2015] [Indexed: 12/24/2022] Open
Abstract
The infiltration of immune cells in the central nervous system is a common hallmark in different neuroinflammatory conditions. Accumulating evidence indicates that resident glial cells can establish a cross-talk with infiltrated immune cells, including T-cells, regulating their recruitment, activation and function within the CNS. Although the healthy CNS has been thought to be devoid of professional dendritic cells (DCs), numerous studies have reported the presence of a population of DCs in specific locations such as the meninges, choroid plexuses and the perivascular space. Moreover, the infiltration of DC precursors during neuroinflammatory situations has been proposed, suggesting a putative role of these cells in the regulation of lymphocyte activity within the CNS. On the other hand, under specific circumstances, microglial cells are able to acquire a phenotype of DC expressing a wide range of molecules that equip these cells with all the necessary machinery for communication with T-cells. In this review, we summarize the current knowledge on the expression of molecules involved in the cross-talk with T-cells in both microglial cells and DCs and discuss the potential contribution of each of these cell populations on the control of lymphocyte function within the CNS.
Collapse
Affiliation(s)
- Beatriz Almolda
- Department of Cell Biology, Physiology and Immunology, Facultat de Medicina, Institute of Neurosciences, Universitat Autònoma de Barcelona Bellaterra, Spain
| | - Berta González
- Department of Cell Biology, Physiology and Immunology, Facultat de Medicina, Institute of Neurosciences, Universitat Autònoma de Barcelona Bellaterra, Spain
| | - Bernardo Castellano
- Department of Cell Biology, Physiology and Immunology, Facultat de Medicina, Institute of Neurosciences, Universitat Autònoma de Barcelona Bellaterra, Spain
| |
Collapse
|
175
|
Hucke S, Eschborn M, Liebmann M, Herold M, Freise N, Engbers A, Ehling P, Meuth SG, Roth J, Kuhlmann T, Wiendl H, Klotz L. Sodium chloride promotes pro-inflammatory macrophage polarization thereby aggravating CNS autoimmunity. J Autoimmun 2015; 67:90-101. [PMID: 26584738 DOI: 10.1016/j.jaut.2015.11.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 10/23/2015] [Accepted: 11/02/2015] [Indexed: 12/13/2022]
Abstract
The increasing incidence in Multiple Sclerosis (MS) during the last decades in industrialized countries might be linked to a change in dietary habits. Nowadays, enhanced salt content is an important characteristic of Western diet and increased dietary salt (NaCl) intake promotes pathogenic T cell responses contributing to central nervous system (CNS) autoimmunity. Given the importance of macrophage responses for CNS disease propagation, we addressed the influence of salt consumption on macrophage responses in CNS autoimmunity. We observed that EAE-diseased mice receiving a NaCl-high diet showed strongly enhanced macrophage infiltration and activation within the CNS accompanied by disease aggravation during the effector phase of EAE. NaCl treatment of macrophages elicited a strong pro-inflammatory phenotype characterized by enhanced pro-inflammatory cytokine production, increased expression of immune-stimulatory molecules, and an antigen-independent boost of T cell proliferation. This NaCl-induced pro-inflammatory macrophage phenotype was accompanied by increased activation of NF-kB and MAPK signaling pathways. The pathogenic relevance of NaCl-conditioned macrophages is illustrated by the finding that transfer into EAE-diseased animals resulted in significant disease aggravation compared to untreated macrophages. Importantly, also in human monocytes, NaCl promoted a pro-inflammatory phenotype that enhanced human T cell proliferation. Taken together, high dietary salt intake promotes pro-inflammatory macrophages that aggravate CNS autoimmunity. Together with other studies, these results underline the need to further determine the relevance of increased dietary salt intake for MS disease severity.
Collapse
Affiliation(s)
| | | | - Marie Liebmann
- Department of Neurology, University of Muenster, Germany
| | - Martin Herold
- Department of Neurology, University of Muenster, Germany
| | - Nicole Freise
- Institute of Immunology, University of Muenster, Germany
| | - Annika Engbers
- Department of Neurology, University of Muenster, Germany
| | - Petra Ehling
- Department of Neurology, University of Muenster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Muenster, Germany; Cells in Motion, Cluster of Excellence, University of Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Muenster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University of Muenster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Muenster, Germany; Cells in Motion, Cluster of Excellence, University of Münster, Germany
| | - Luisa Klotz
- Department of Neurology, University of Muenster, Germany.
| |
Collapse
|
176
|
Di Dario M, Colombo E, Govi C, De Feo D, Messina MJ, Romeo M, Sangalli F, Moiola L, Rodegher M, Martino G, Martinelli V, Comi G, Farina C. Myeloid cells as target of fingolimod action in multiple sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e157. [PMID: 26587553 PMCID: PMC4635549 DOI: 10.1212/nxi.0000000000000157] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 08/05/2015] [Indexed: 01/03/2023]
Abstract
Objective: To track the effects of fingolimod, an approved drug for multiple sclerosis (MS), on the activation of myeloid cells from the periphery to the CNS. Methods: In vitro and ex vivo immunologic studies coupled with flow cytometry were performed to evaluate the action of fingolimod on lipopolysaccharide (LPS)–induced expression of activation markers in human monocytes from healthy participants, participants with untreated MS, and participants with fingolimod-treated MS. In vivo administration of fingolimod during experimental autoimmune encephalomyelitis (EAE) was established to verify the activation state of splenic, CNS infiltrating, and CNS resident myeloid cells ex vivo at flow cytometer. Results: We found that in vitro exposure of human monocytes to fingolimod inhibited LPS-induced CD25 and CD150 expression and tumor necrosis factor–α (TNF-α) secretion without altering immune cell survival. Further, EAE treatment with fingolimod led to reduced amounts of TNF-α produced by myeloid cells in vivo in the spleen and CNS. Finally, while displaying normal induction of CD25 and CD150 levels at high LPS concentration, monocytes from patients with fingolimod-treated MS showed significantly higher activation threshold at suboptimal LPS stimulation than controls. Conclusions: The inhibition of myeloid cell activation may be part of the immunosuppressive action of fingolimod and take place in the periphery and in the CNS.
Collapse
Affiliation(s)
- Marco Di Dario
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuela Colombo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Govi
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Donatella De Feo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria José Messina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marzia Romeo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sangalli
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mariaemma Rodegher
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianvito Martino
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vittorio Martinelli
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giancarlo Comi
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
177
|
Shaked I, Hanna RN, Shaked H, Chodaczek G, Nowyhed HN, Tweet G, Tacke R, Basat AB, Mikulski Z, Togher S, Miller J, Blatchley A, Salek-Ardakani S, Darvas M, Kaikkonen MU, Thomas GD, Lai-Wing-Sun S, Rezk A, Bar-Or A, Glass CK, Bandukwala H, Hedrick CC. Transcription factor Nr4a1 couples sympathetic and inflammatory cues in CNS-recruited macrophages to limit neuroinflammation. Nat Immunol 2015; 16:1228-34. [PMID: 26523867 DOI: 10.1038/ni.3321] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022]
Abstract
The molecular mechanisms that link the sympathetic stress response and inflammation remain obscure. Here we found that the transcription factor Nr4a1 regulated the production of norepinephrine (NE) in macrophages and thereby limited experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. Lack of Nr4a1 in myeloid cells led to enhanced NE production, accelerated infiltration of leukocytes into the central nervous system (CNS) and disease exacerbation in vivo. In contrast, myeloid-specific deletion of tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis, protected mice against EAE. Furthermore, we found that Nr4a1 repressed autocrine NE production in macrophages by recruiting the corepressor CoREST to the Th promoter. Our data reveal a new role for macrophages in neuroinflammation and identify Nr4a1 as a key regulator of catecholamine production by macrophages.
Collapse
Affiliation(s)
- Iftach Shaked
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Richard N Hanna
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Helena Shaked
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Grzegorz Chodaczek
- Microscopy Core, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Heba N Nowyhed
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - George Tweet
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Robert Tacke
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Alp Bugra Basat
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Zbigniew Mikulski
- Microscopy Core, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Susan Togher
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Jacqueline Miller
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Amy Blatchley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Shahram Salek-Ardakani
- Department of Pathology, Immunology &Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Martin Darvas
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Minna U Kaikkonen
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Graham D Thomas
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | | | - Ayman Rezk
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Amit Bar-Or
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Christopher K Glass
- Department of Cellular &Molecular Medicine, University of California San Diego, San Diego, California, USA
| | - Hozefa Bandukwala
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| |
Collapse
|
178
|
Melero-Jerez C, Ortega MC, Moliné-Velázquez V, Clemente D. Myeloid derived suppressor cells in inflammatory conditions of the central nervous system. Biochim Biophys Acta Mol Basis Dis 2015; 1862:368-80. [PMID: 26527182 DOI: 10.1016/j.bbadis.2015.10.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/16/2015] [Accepted: 10/19/2015] [Indexed: 12/11/2022]
Abstract
The knowledge of the immune system elements and their relationship with other tissues, organs and systems are key approximations for the resolution of many immune-related disorders. The control of the immune response and/or its modulation from the pro-inflammatory to the anti-inflammatory response is being deeply studied in the field. In the last years, the study of myeloid-derived suppressor cells (MDSCs), a group of immature myeloid cells with a high suppressive activity on T cells has been extensively addressed in cancer. In contrast, their role in neuroimmune diseases is far from being totally understood. In this review, we will summarize data about MDSCs coming from the study of neuroinflammatory diseases in general and their potential role in multiple sclerosis, in order to introduce the putative use of this extraordinary promising cell type for future cell-based therapies. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
Affiliation(s)
- Carolina Melero-Jerez
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda" s/n, E-45071 Toledo, Spain
| | - María Cristina Ortega
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda" s/n, E-45071 Toledo, Spain; Centro de Biología Molecular Severo Ochoa. Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Verónica Moliné-Velázquez
- Animal Experimental Unit, Scientific Instrumentation Center (CIC), Campus de la Cartuja, Universidad de Granada, Granada, Spain
| | - Diego Clemente
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda" s/n, E-45071 Toledo, Spain.
| |
Collapse
|
179
|
Wieghofer P, Prinz M. Genetic manipulation of microglia during brain development and disease. Biochim Biophys Acta Mol Basis Dis 2015; 1862:299-309. [PMID: 26432479 DOI: 10.1016/j.bbadis.2015.09.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 09/24/2015] [Accepted: 09/25/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Peter Wieghofer
- Institute of Neuropathology, University of Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Germany.
| |
Collapse
|
180
|
Larochelle A, Bellavance MA, Michaud JP, Rivest S. Bone marrow-derived macrophages and the CNS: An update on the use of experimental chimeric mouse models and bone marrow transplantation in neurological disorders. Biochim Biophys Acta Mol Basis Dis 2015; 1862:310-22. [PMID: 26432480 DOI: 10.1016/j.bbadis.2015.09.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 09/17/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022]
Abstract
The central nervous system (CNS) is a very unique system with multiple features that differentiate it from systemic tissues. One of the most captivating aspects of its distinctive nature is the presence of the blood brain barrier (BBB), which seals it from the periphery. Therefore, to preserve tissue homeostasis, the CNS has to rely heavily on resident cells such as microglia. These pivotal cells of the mononuclear lineage have important and dichotomous roles according to various neurological disorders. However, certain insults can overwhelm microglia as well as compromising the integrity of the BBB, thus allowing the infiltration of bone marrow-derived macrophages (BMDMs). The use of myeloablation and bone marrow transplantation allowed the generation of chimeric mice to study resident microglia and infiltrated BMDM separately. This breakthrough completely revolutionized the way we captured these 2 types of mononuclear phagocytic cells. We now realize that microglia and BMDM exhibit distinct features and appear to perform different tasks. Since these cells are central in several pathologies, it is crucial to use chimeric mice to analyze their functions and mechanisms to possibly harness them for therapeutic purpose. This review will shed light on the advent of this methodology and how it allowed deciphering the ontology of microglia and its maintenance during adulthood. We will also compare the different strategies used to perform myeloablation. Finally, we will discuss the landmark studies that used chimeric mice to characterize the roles of microglia and BMDM in several neurological disorders. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
Affiliation(s)
- Antoine Larochelle
- Neuroscience Laboratory, CHU de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada
| | - Marc-André Bellavance
- Neuroscience Laboratory, CHU de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada
| | - Jean-Philippe Michaud
- Neuroscience Laboratory, CHU de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center, Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Blvd., Québec G1V 4G2, Canada.
| |
Collapse
|
181
|
Stojić-Vukanić Z, Nacka-Aleksić M, Pilipović I, Vujnović I, Blagojević V, Kosec D, Dimitrijević M, Leposavić G. Aging diminishes the resistance of AO rats to EAE: putative role of enhanced generation of GM-CSF Expressing CD4+ T cells in aged rats. IMMUNITY & AGEING 2015; 12:16. [PMID: 26448779 PMCID: PMC4596406 DOI: 10.1186/s12979-015-0044-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Aging influences immune response and susceptibility to EAE in a strain specific manner. The study was designed to examine influence of aging on EAE induction in Albino Oxford (AO) rats. RESULTS Differently from 3-month-old (young) rats, which were resistant to EAE induction, the majority of aged (24-26-month-old) rats developed mild chronic form of EAE. On 16(th) day post-immunization, when in aged rats the neurological deficit reached plateau, more mononuclear cells, including CD4+ T lymphocytes was retrieved from spinal cord of aged than young rats. The frequencies of IL-17+ and GM-CSF+ cells within spinal cord infiltrating CD4+ lymphocytes were greater in aged rats. To their increased frequency contributed the expansion of GM-CSF + IL-17 + IFN-γ+ cells, which are highly pathogenic in mice. The expression of the cytokines (IL-1β and IL-23/p19) driving GM-CSF + IL-17 + IFN-γ + cell differentiation in mice was also augmented in aged rat spinal cord mononuclear cells. Additionally, in aged rat spinal cord the expansion of GM-CSF + IL-17-IFN-γ- CD4+ T lymphocytes was found. Consistently, the expression of mRNAs for IL-3, the cytokine exhibiting the same expression pattern as GM-CSF, and IL-7, the cytokine driving differentiation of GM-CSF + IL-17-IFN-γ- CD4 + lymphocytes in mice, was upregulated in aged rat spinal cord mononuclear cells, and the tissue, respectively. This was in accordance with the enhanced generation of the brain antigen-specific GM-CSF+ CD4+ lymphocytes in aged rat draining lymph nodes, as suggested by (i) the higher frequency of GM-CSF+ cells (reflecting the expansion of IL-17-IFN-γ- cells) within their CD4+ lymphocytes and (ii) the upregulated GM-CSF and IL-3 mRNA expression in fresh CD4+ lymphocytes and MBP-stimulated draining lymph node cells and IL-7 mRNA in lymph node tissue from aged rats. In agreement with the upregulated GM-CSF expression in aged rats, strikingly more CD11b + CD45(int) (activated microglia) and CD45(hi) (mainly proinflammatory dendritic cells and macrophages) cells was retrieved from aged than young rat spinal cord. Besides, expression of mRNA for SOCS1, a negative regulator of proinflammatory cytokine expression in innate immunity cells, was downregulated in aged rat spinal cord mononuclear cells. CONCLUSIONS The study revealed that aging may overcome genetic resistance to EAE, and indicated the cellular and molecular mechanisms contributing to this phenomenon in AO rats.
Collapse
Affiliation(s)
- Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Nacka-Aleksić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivana Vujnović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Veljko Blagojević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Mirjana Dimitrijević
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| |
Collapse
|
182
|
Meyer-Luehmann M, Prinz M. Myeloid Cells in Alzheimer's Disease: Culprits, Victims or Innocent Bystanders? Trends Neurosci 2015; 38:659-668. [DOI: 10.1016/j.tins.2015.08.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 08/30/2015] [Accepted: 08/31/2015] [Indexed: 12/23/2022]
|
183
|
Interleukin-36γ is expressed by neutrophils and can activate microglia, but has no role in experimental autoimmune encephalomyelitis. J Neuroinflammation 2015; 12:173. [PMID: 26377915 PMCID: PMC4574267 DOI: 10.1186/s12974-015-0392-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/03/2015] [Indexed: 12/25/2022] Open
Abstract
Background Experimental autoimmune encephalomyelitis (EAE) is a model of inflammatory demyelinating diseases mediated by different types of leukocytes. How these cells communicate with each other to orchestrate autoimmune attacks is not fully understood, especially in the case of neutrophils, whose importance in EAE is newly established. The present study aimed to determine the expression pattern and role of different components of the IL-36 signaling pathway (IL-36α, IL-36β, IL-36γ, IL-36R) in EAE. Methods EAE was induced by either active immunization with myelin peptide, passive transfer of myelin-reactive T cells or injection of pertussis toxin to transgenic 2D2 mice. The molecules of interest were analyzed using a combination of techniques, including quantitative real-time PCR (qRT-PCR), flow cytometry, Western blotting, in situ hybridization, and immunohistochemistry. Microglial cultures were treated with recombinant IL-36γ and analyzed using DNA microarrays. Different mouse strains were subjected to clinical evaluation and flow cytometric analysis in order to compare their susceptibility to EAE. Results Our observations indicate that both IL-36γ and IL-36R are strongly upregulated in nervous and hematopoietic tissues in different forms of EAE. IL-36γ is specifically expressed by neutrophils, while IL-36R is expressed by different immune cells, including microglia and other myeloid cells. In culture, microglia respond to recombinant IL-36γ by expressing molecules involved in neutrophil recruitment, such as Csf3, IL-1β, and Cxcl2. However, mice deficient in either IL-36γ or IL-36R develop similar clinical and histopathological signs of EAE compared to wild-type controls. Conclusion This study identifies IL-36γ as a neutrophil-related cytokine that can potentially activate microglia, but that is only correlative and not contributory in EAE. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0392-7) contains supplementary material, which is available to authorized users.
Collapse
|
184
|
Grigoriadis N, van Pesch V. A basic overview of multiple sclerosis immunopathology. Eur J Neurol 2015; 22 Suppl 2:3-13. [DOI: 10.1111/ene.12798] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 06/05/2015] [Indexed: 12/12/2022]
Affiliation(s)
- N. Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology; Second Department of Neurology; AHEPA University Hospital; Aristotle University of Thessaloniki; Macedonia Greece
| | - V. van Pesch
- Neurology Department; Cliniques Universitaires St-Luc; Brussels Belgium
| | | |
Collapse
|
185
|
Croxford A, Lanzinger M, Hartmann F, Schreiner B, Mair F, Pelczar P, Clausen B, Jung S, Greter M, Becher B. The Cytokine GM-CSF Drives the Inflammatory Signature of CCR2+ Monocytes and Licenses Autoimmunity. Immunity 2015; 43:502-14. [DOI: 10.1016/j.immuni.2015.08.010] [Citation(s) in RCA: 314] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/04/2015] [Accepted: 06/01/2015] [Indexed: 11/28/2022]
|
186
|
Do not judge a cell by its cover--diversity of CNS resident, adjoining and infiltrating myeloid cells in inflammation. Semin Immunopathol 2015; 37:591-605. [PMID: 26251238 DOI: 10.1007/s00281-015-0520-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/27/2015] [Indexed: 12/24/2022]
Abstract
Specialized populations of tissue-resident myeloid cells inhabit every organ of the body. While many of these populations appear similar morphologically and phenotypically, they exhibit great functional diversity. The central nervous system (CNS), as an immune privileged organ, possesses a unique tissue-resident macrophage population, the microglia, as well as numerous myeloid cell subsets at its boarders and barriers in CNS-adjoining tissues, namely the meninges, the perivascular space, and the choroid plexus. Recent research has added much to our knowledge about microglia, whereas the populations of CNS-surrounding phagocytes are just starting to be appreciated. As guardians of CNS homeostasis, these myeloid cells perform immune surveillance and immune modulatory tasks in health and disease. As such, microglia and CNS-surrounding antigen-presenting cells have been shown to be crucially involved not only in the initiation and progression but also resolution of multiple sclerosis (MS). MS and its rodent model, experimental autoimmune encephalomyelitis, are autoimmune inflammatory demyelinating CNS pathologies. While some crucial aspects of the disease pathogenesis have been solved, much of the complex involvement and interplay of the innate immune compartment remains yet to be clarified. Here, we will discuss the current understanding of the scope of phenotypes and functions of myeloid cells involved in CNS neuroinflammation.
Collapse
|
187
|
Ginhoux F, Prinz M. Origin of microglia: current concepts and past controversies. Cold Spring Harb Perspect Biol 2015; 7:a020537. [PMID: 26134003 DOI: 10.1101/cshperspect.a020537] [Citation(s) in RCA: 279] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS), which sit in close proximity to neural structures and are intimately involved in brain homeostasis. The microglial population also plays fundamental roles during neuronal expansion and differentiation, as well as in the perinatal establishment of synaptic circuits. Any change in the normal brain environment results in microglial activation, which can be detrimental if not appropriately regulated. Aberrant microglial function has been linked to the development of several neurological and psychiatric diseases. However, microglia also possess potent immunoregulatory and regenerative capacities, making them attractive targets for therapeutic manipulation. Such rationale manipulations will, however, require in-depth knowledge of their origins and the molecular mechanisms underlying their homeostasis. Here, we discuss the latest advances in our understanding of the origin, differentiation, and homeostasis of microglial cells and their myelomonocytic relatives in the CNS.
Collapse
Affiliation(s)
- Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, 79106 Freiburg, Germany BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
188
|
Yang H, Bi Y, Han F, Lu Y, Wang J, Zhang Z, Liu G. Myeloid-derived suppressor cells in immunity and autoimmunity. Expert Rev Clin Immunol 2015; 11:911-9. [DOI: 10.1586/1744666x.2015.1052794] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
189
|
Greter M, Lelios I, Croxford AL. Microglia Versus Myeloid Cell Nomenclature during Brain Inflammation. Front Immunol 2015; 6:249. [PMID: 26074918 PMCID: PMC4443742 DOI: 10.3389/fimmu.2015.00249] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/07/2015] [Indexed: 12/05/2022] Open
Abstract
As immune sentinels of the central nervous system (CNS), microglia not only respond rapidly to pathological conditions but also contribute to homeostasis in the healthy brain. In contrast to other populations of the myeloid lineage, adult microglia derive from primitive myeloid precursors that arise in the yolk sac early during embryonic development, after which they self-maintain locally and independently of blood-borne myeloid precursors. Under neuro-inflammatory conditions such as experimental autoimmune encephalomyelitis, circulating monocytes invade the CNS parenchyma where they further differentiate into macrophages or inflammatory dendritic cells. Often it is difficult to delineate resident microglia from infiltrating myeloid cells using currently known markers. Here, we will discuss the current means to reliably distinguish between these populations, and which recent advances have helped to make clear definitions between phenotypically similar, yet functionally diverse myeloid cell types.
Collapse
Affiliation(s)
- Melanie Greter
- Institute of Experimental Immunology, University of Zurich , Zurich , Switzerland
| | - Iva Lelios
- Institute of Experimental Immunology, University of Zurich , Zurich , Switzerland
| | | |
Collapse
|
190
|
Morris DL. Minireview: Emerging Concepts in Islet Macrophage Biology in Type 2 Diabetes. Mol Endocrinol 2015; 29:946-62. [PMID: 26001058 DOI: 10.1210/me.2014-1393] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Chronic systemic inflammation is a hallmark feature of obesity and type 2 diabetes. Both resident and recruited islet macrophages contribute to the proinflammatory milieu of the diabetic islet. However, macrophages also appear to be critical for β-cell formation during development and support β-cell replication in experimental models of pancreas regeneration. In light of these findings, perhaps macrophages in the islet need to be viewed more as a fulcrum where deleterious inflammatory activation is balanced with beneficial tissue repair processes. Undoubtedly, defining the factors that contribute to the ontogeny, heterogeneity, and functionality of macrophages in normal, diseased, and regenerating islets will be necessary to determine whether that fulcrum can be moved to preserve functional β-cell mass in persons with diabetes. The intent of this review is to introduce the reader to emerging concepts of islet macrophage biology that may challenge the perception that macrophage accumulation in islets is merely a pathological feature of type 2 diabetes.
Collapse
Affiliation(s)
- David L Morris
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
191
|
Constantinescu CS, Asher A, Fryze W, Kozubski W, Wagner F, Aram J, Tanasescu R, Korolkiewicz RP, Dirnberger-Hertweck M, Steidl S, Libretto SE, Sprenger T, Radue EW. Randomized phase 1b trial of MOR103, a human antibody to GM-CSF, in multiple sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e117. [PMID: 26185773 PMCID: PMC4442097 DOI: 10.1212/nxi.0000000000000117] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/22/2015] [Indexed: 11/15/2022]
Abstract
Objectives: To determine the safety, pharmacokinetics (PK), and immunogenicity of the recombinant human monoclonal antibody MOR103 to granulocyte-macrophage colony-stimulating factor (GM-CSF) in patients with multiple sclerosis (MS) with clinical or MRI activity. Methods: In this 20-week, randomized, double-blind, placebo-controlled phase 1b dose-escalation trial (registration number NCT01517282), adults with relapsing-remitting MS (RRMS) or secondary progressive MS (SPMS) received an IV infusion of placebo (n = 6) or MOR103 0.5 (n = 8), 1.0 (n = 8), or 2.0 (n = 9) mg/kg every 2 weeks for 10 weeks. Patients had to have ≤10 gadolinium (Gd)-enhancing brain lesions on T1-weighted MRI at baseline. The primary objective was safety. Results: Most treatment-emergent adverse events (TEAEs) were mild to moderate in severity. The most frequent was nasopharyngitis. Between-group differences in TEAE numbers were small. There were no TEAE-related trial discontinuations, infusion-related reactions, or deaths. Nine patients experienced MS exacerbations: 3, 5, 1, and 0 patient(s) in the placebo, 0.5, 1.0, and 2.0 mg/kg groups, respectively. A few T1 Gd-enhancing lesions and/or new or enlarging T2 lesions indicative of inflammation were observed in all treatment groups. No clinically significant changes were observed in other clinical assessments or laboratory safety assessments. No anti-MOR103 antibodies were detected. PK evaluations indicated dose linearity with low/no drug accumulation over time. Conclusions: MOR103 was generally well-tolerated in patients with RRMS or SPMS. No evidence of immunogenicity was found. Classification of evidence: This phase 1b study provides Class I evidence that MOR103 has acceptable tolerability in patients with MS.
Collapse
Affiliation(s)
- Cris S Constantinescu
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Aliya Asher
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Waldemar Fryze
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Wojciech Kozubski
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Frank Wagner
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Jehan Aram
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Radu Tanasescu
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Roman P Korolkiewicz
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Maren Dirnberger-Hertweck
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Stefan Steidl
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Susan E Libretto
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Till Sprenger
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| | - Ernst W Radue
- Division of Clinical Neuroscience (C.S.C., J.A., R.T.), Research Group in Clinical Neurology, University of Nottingham, Queen's Medical Centre, Nottingham, UK; MAC Clinical Research Ltd. (A.A.), Trafford Park, Manchester, UK; Pomorskie Centrum Traumatologii im. M. Kopernika w Gdańsku (W.F.), Oddzial Neurologiczny i Udarowy, Gdańsk, Poland; Oddział Kliniczny Neurologii (W.K.), Medical University of Poznan, Poznań, Poland; Charité Research Organisation GmbH (F.W.), Berlin, Germany; MorphoSys AG (R.P.K., M.D.-H., S.S.), Martinsried/Planegg, Germany; Sue Libretto Publications Consultant Ltd. (S.E.L.), Windlesham, Surrey, UK; and MIAC AG (T.S., E.W.R.) and Department of Neurology (T.S.), University Hospital Basel, Basel, Switzerland
| |
Collapse
|
192
|
Kwak Y, Kim HE, Park SG. Insights into Myeloid-Derived Suppressor Cells in Inflammatory Diseases. Arch Immunol Ther Exp (Warsz) 2015; 63:269-85. [PMID: 25990434 DOI: 10.1007/s00005-015-0342-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/14/2015] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells involved in immune regulation. This population subdivides into granulocytic MDSCs and monocytic MDSCs, which regulate immune responses via the production of various molecules including reactive oxygen species, nitric oxide, arginase-1, interleukin-10, and transforming growth factor-β. Most studies of MDSCs focused on their role in tumors. MDSCs protect tumor cells from immune responses, and thus the frequency of MDSCs associates with poor prognosis. Many recent studies reported an important role for MDSCs in inflammatory diseases via the regulation of immune cells. In addition, the utilization of MDSCs by infectious pathogens suggests an immune evasion mechanism. Thus, MDSCs are important immune regulators in inflammatory diseases, as well as in tumors. This review focuses on the role of MDSCs in the regulation of inflammation in non-tumor settings.
Collapse
Affiliation(s)
- Yewon Kwak
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 500-712, Republic of Korea
| | | | | |
Collapse
|
193
|
The Role and Potential Therapeutic Application of Myeloid-Derived Suppressor Cells in Allo- and Autoimmunity. Mediators Inflamm 2015; 2015:421927. [PMID: 26078493 PMCID: PMC4452474 DOI: 10.1155/2015/421927] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/21/2015] [Accepted: 04/28/2015] [Indexed: 12/16/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that consists of myeloid progenitor cells and immature myeloid cells. They have been identified as a cell population that may affect the activation of CD4(+) and CD8(+) T-cells to regulate the immune response negatively, which makes them attractive targets for the treatment of transplantation and autoimmune diseases. Several studies have suggested the potential suppressive effect of MDSCs on allo- and autoimmune responses. Conversely, MDSCs have also been found at various stages of differentiation, accumulating during pathological situations, not only during tumor development but also in a variety of inflammatory immune responses, bone marrow transplantation, and some autoimmune diseases. These findings appear to be contradictory. In this review, we summarize the roles of MDSCs in different transplantation and autoimmune diseases models as well as the potential to target these cells for therapeutic benefit.
Collapse
|
194
|
Abou-Hamdan M, Costanza M, Fontana E, Di Dario M, Musio S, Congiu C, Onnis V, Lattanzi R, Radaelli M, Martinelli V, Salvadori S, Negri L, Poliani PL, Farina C, Balboni G, Steinman L, Pedotti R. Critical role for prokineticin 2 in CNS autoimmunity. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e95. [PMID: 25884014 PMCID: PMC4396530 DOI: 10.1212/nxi.0000000000000095] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/02/2015] [Indexed: 12/14/2022]
Abstract
Objective: To investigate the potential role of prokineticin 2 (PK2), a bioactive peptide involved in multiple biological functions including immune modulation, in CNS autoimmune demyelinating disease. Methods: We investigated the expression of PK2 in mice with experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis (MS), and in patients with relapsing-remitting MS. We evaluated the biological effects of PK2 on expression of EAE and on development of T-cell response against myelin by blocking PK2 in vivo with PK2 receptor antagonists. We treated with PK2 immune cells activated against myelin antigen to explore the immune-modulating effects of this peptide in vitro. Results: Pk2 messenger RNA was upregulated in spinal cord and lymph node cells (LNCs) of mice with EAE. PK2 protein was expressed in EAE inflammatory infiltrates and was increased in sera during EAE. In patients with relapsing-remitting MS, transcripts for PK2 were significantly increased in peripheral blood mononuclear cells compared with healthy controls, and PK2 serum concentrations were significantly higher. A PK2 receptor antagonist prevented or attenuated established EAE in chronic and relapsing-remitting models, reduced CNS inflammation and demyelination, and decreased the production of interferon (IFN)-γ and interleukin (IL)-17A cytokines in LNCs while increasing IL-10. PK2 in vitro increased IFN-γ and IL-17A and reduced IL-10 in splenocytes activated against myelin antigen. Conclusion: These data suggest that PK2 is a critical immune regulator in CNS autoimmune demyelination and may represent a new target for therapy.
Collapse
Affiliation(s)
- Mhamad Abou-Hamdan
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Massimo Costanza
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Elena Fontana
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Marco Di Dario
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Silvia Musio
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Cenzo Congiu
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Valentina Onnis
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Roberta Lattanzi
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Marta Radaelli
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Vittorio Martinelli
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Severo Salvadori
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Lucia Negri
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Pietro Luigi Poliani
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Cinthia Farina
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Gianfranco Balboni
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Lawrence Steinman
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| | - Rosetta Pedotti
- Neuroimmunology and Neuromuscular Disorder Unit (M.A.-H., M.C., S.M., R.P.), Neurological Institute Foundation IRCCS Carlo Besta, Milan, Italy; Department of Molecular and Translational Medicine (E.F., P.L.P.), Pathology Unit, University of Brescia, Italy; Institute of Experimental Neurology (M.D.D., M.R., V.M., C.F.), San Raffaele Scientific Institute, Milan, Italy; Department of Life and Environmental Sciences (C.C., V.O., G.B.), Pharmaceutical, Pharmacological and Nutraceutical Sciences Unit, University of Cagliari, Italy; Department of Physiology and Pharmacology Vittorio Erspamer (R.L., L.N.), Sapienza University of Rome, Italy; Department of Chemical and Pharmaceutical Sciences (S.S.), University of Ferrara, Italy; and Department of Neurology (L.S., R.P.), Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
195
|
CCR2 antagonism alters brain macrophage polarization and ameliorates cognitive dysfunction induced by traumatic brain injury. J Neurosci 2015; 35:748-60. [PMID: 25589768 DOI: 10.1523/jneurosci.2405-14.2015] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Traumatic brain injury (TBI) is a major risk factor for the development of multiple neurodegenerative diseases. With respect to the increasing prevalence of TBI, new therapeutic strategies are urgently needed that will prevent secondary damage to primarily unaffected tissue. Consistently, neuroinflammation has been implicated as a key mediator of secondary damage following the initial mechanical insult. Following injury, there is uncertainty regarding the role that accumulating CCR2(+) macrophages play in the injury-induced neuroinflammatory sequelae and cognitive dysfunction. Using CX3CR1(GFP/+)CCR2(RFP/+) reporter mice, we show that TBI initiated a temporally restricted accumulation of peripherally derived CCR2(+) macrophages, which were concentrated in the hippocampal formation, a region necessary for learning and memory. Multivariate analysis delineated CCR2(+) macrophages' neuroinflammatory response while identifying a novel therapeutic treatment window. As a proof of concept, targeting CCR2(+) macrophages with CCX872, a novel Phase I CCR2 selective antagonist, significantly reduced TBI-induced inflammatory macrophage accumulation. Concomitantly, there was a significant reduction in multiple proinflammatory and neurotoxic mediators with this treatment paradigm. Importantly, CCR2 antagonism resulted in a sparing of TBI-induced hippocampal-dependent cognitive dysfunction and reduced proinflammatory activation profile 1 month after injury. Thus, therapeutically targeting the CCR2(+) subset of monocytes/macrophages may provide a new avenue of clinical intervention following TBI.
Collapse
|
196
|
Pivotal roles of GM-CSF in autoimmunity and inflammation. Mediators Inflamm 2015; 2015:568543. [PMID: 25838639 PMCID: PMC4370199 DOI: 10.1155/2015/568543] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/23/2015] [Indexed: 12/14/2022] Open
Abstract
Granulocyte macrophage-colony stimulating factor (GM-CSF) is a hematopoietic growth factor, which stimulates the proliferation of granulocytes and macrophages from bone marrow precursor cells. In autoimmune and inflammatory diseases, Th17 cells have been considered as strong inducers of tissue inflammation. However, recent evidence indicates that GM-CSF has prominent proinflammatory functions and that this growth factor (not IL-17) is critical for the pathogenicity of CD4+ T cells. Therefore, the mechanism of GM-CSF-producing CD4+ T cell differentiation and the role of GM-CSF in the development of autoimmune and inflammatory diseases are gaining increasing attention. This review summarizes the latest knowledge of GM-CSF and its relationship with autoimmune and inflammatory diseases. The potential therapies targeting GM-CSF as well as their possible side effects have also been addressed in this review.
Collapse
|
197
|
Zhang H, Wang S, Huang Y, Wang H, Zhao J, Gaskin F, Yang N, Fu SM. Myeloid-derived suppressor cells are proinflammatory and regulate collagen-induced arthritis through manipulating Th17 cell differentiation. Clin Immunol 2015; 157:175-86. [PMID: 25680967 DOI: 10.1016/j.clim.2015.02.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 02/01/2015] [Accepted: 02/03/2015] [Indexed: 12/12/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) and Th17 cells were found to expand in collagen-induced arthritis (CIA) significantly. Two subsets of MDSC, polymorphonuclear (PMN) and mononuclear (MO), were detected and their ratios varied during the development of CIA. The depletion of MDSC in vivo resulted in suppression of T-cell proliferation and decreased IL-17A and IL-1β production. The adoptive transfer of MDSC restored the severity of arthritis and Th17 cell differentiation. The depletion of MDSCs on day 35 resulted in arthritis amelioration without reaching a significant difference. Furthermore, MDSCs from CIA mice had higher production of IL-1β and promoted Th17 cell differentiation. The expansion of MDSCs in the peripheral blood of rheumatoid arthritis (RA) patients was in correlation with increased Th17 cells and disease activity DAS28. These results support the hypothesis that MDSC may play a significant proinflammatory role in the pathogenesis of CIA and RA by inducing Th17 development in an IL-1β-dependent manner.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Shuang Wang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Yuefang Huang
- Department of Pediatrics, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, PR China
| | - Hongyue Wang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Jijun Zhao
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Felicia Gaskin
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22908-0133, USA
| | - Niansheng Yang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China.
| | - Shu Man Fu
- Division of Rheumatology and Center of inflammation, Immunology and Regenerative Medicine, Department of Medicine and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908-0133, USA
| |
Collapse
|
198
|
Galvani RG, Lemos R, Areal RB, Salvador PA, Zamboni DS, Wanderley JLM, Bonomo A. Disease severity and mortality can be independently regulated in a mouse model of experimental graft versus host disease. PLoS One 2015; 10:e0118079. [PMID: 25643148 PMCID: PMC4313938 DOI: 10.1371/journal.pone.0118079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 01/05/2015] [Indexed: 12/16/2022] Open
Abstract
Graft versus host disease (GVHD) is the major limitation of allogeneic hematopoietic stem cell transplantation (HSCT) presenting high mortality and morbidity rates. However, the exact cause of death is not completely understood and does not correlate with specific clinical and histological parameters of disease. Here we show, by using a semi-allogeneic mouse model of GVHD, that mortality and morbidity can be experimentally separated. We injected bone marrow-derived dendritic cells (BMDC) from NOD2/CARD15-deficient donors into semi-allogeneic irradiated chimaeras and observed that recipients were protected from death. However, no protection was observed regarding clinical or pathological scores up to 20 days after transplantation. Protection from death was associated with decreased bacterial translocation, faster hematologic recovery and epithelial integrity maintenance despite mononuclear infiltration at day 20 post-GVHD induction with no skew towards different T helper phenotypes. The protected mice recovered from aGVHD and progressively reached scores compatible with healthy animals. Altogether, our data indicate that severity and mortality can be separate events providing a model to study transplant-related mortality.
Collapse
Affiliation(s)
- Rômulo G. Galvani
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ramon Lemos
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rômulo B. Areal
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Pollyanna A. Salvador
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Dario S. Zamboni
- Departamento de Biologia Celular, Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - João Luiz M. Wanderley
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- NUPEM, Campus Macaé Professor Aloísio Teixeira, Universidade Federal do Rio de Janeiro, Macaé, Rio de Janeiro, Brazil
- * E-mail:
| | - Adriana Bonomo
- Divisão de Medicina Experimental, Coordenação de Pesquisa, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratorio de Pesquisa sobre o Timo, Instituo Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
199
|
Du Four S, Maenhout SK, De Pierre K, Renmans D, Niclou SP, Thielemans K, Neyns B, Aerts JL. Axitinib increases the infiltration of immune cells and reduces the suppressive capacity of monocytic MDSCs in an intracranial mouse melanoma model. Oncoimmunology 2015; 4:e998107. [PMID: 26137411 DOI: 10.1080/2162402x.2014.998107] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/08/2014] [Accepted: 12/09/2014] [Indexed: 12/21/2022] Open
Abstract
Melanoma patients are at a high risk of developing brain metastases, which are strongly vascularized and therefore have a significant risk of spontaneous bleeding. VEGF not only plays a role in neo-angiogenesis but also in the antitumor immune response. VEGFR-targeted therapy might not only have an impact on the tumor vascularization but also on tumor-infiltrating immune cells. In this study, we investigated the effect of axitinib, a small molecule TKI of VEGFR-1, -2, and -3, on tumor growth and on the composition of tumor-infiltrating immune cells in subcutaneous and intracranial mouse melanoma models. In vivo treatment with axitinib induced a strong inhibition of tumor growth and significantly improved survival in both tumor models. Characterization of the immune cells within the spleen and tumor of tumor-bearing mice respectively showed a significant increase in the number of CD3+CD8+ T cells and CD11b+ cells of axitinib-treated mice. More specifically, we observed a significant increase of intratumoral monocytic myeloid-derived suppressor cells (moMDSCs; CD11b+Ly6ChighLy6G-). Interestingly, in vitro proliferation assays showed that moMDSCs isolated from spleen or tumor of axitinib-treated mice had a reduced suppressive capacity on a per cell basis as compared to those isolated from vehicle-treated mice. Moreover, MDSCs from axitinib-treated animals displayed the capacity to stimulate allogeneic T cells. Thus, treatment with axitinib induces differentiation of moMDSC toward an antigen-presenting phenotype. Based on these observations, we conclude that the impact of axitinib on tumor growth and survival is most likely not restricted to direct anti-angiogenic effects but also involves important effects on tumor immunity.
Collapse
Key Words
- BLI, bioluminescent imaging
- DCs, Dendritic Cells
- FDA, US Food and Drug Administration
- IL-2, interleukin-2
- MDSC
- MDSC, myeloid-derived suppressor cells
- OT-1, CD8+ T-cells with transgenic receptor specific for the H-2Kb-restricted ovalbumin (OVA) peptide SIINFEKL
- PD-1, programmed death 1
- PD-L1, programmed death 1 ligand
- PFS, progression-free survival
- TKI, Tyrosine Kinase Inhibitor
- TNFα, Tumor Necrosis Factor alfa
- Treg, regulatory T cells
- VEGF, Vascular Endothelial Growth Factor
- angiogenesis
- axitinib
- brain metastasis
- grMDSC, granulocytic MDSC, IFNγ: interferon gamma
- immune cells
- melanoma
- moMDSC, monocytic MDSC
Collapse
Affiliation(s)
- Stephanie Du Four
- Laboratory of Molecular and Cellular Therapy; Department of Immunology-Physiology; Vrije Universiteit Brussel , Brussels, Belgium
| | - Sarah K Maenhout
- Laboratory of Molecular and Cellular Therapy; Department of Immunology-Physiology; Vrije Universiteit Brussel , Brussels, Belgium
| | | | - Dries Renmans
- Laboratory of Molecular and Cellular Therapy; Department of Immunology-Physiology; Vrije Universiteit Brussel , Brussels, Belgium
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory; Luxembourg Institute of Health (LIH) ; Luxembourg
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy; Department of Immunology-Physiology; Vrije Universiteit Brussel , Brussels, Belgium
| | - Bart Neyns
- Department of Medical Oncology ; UZ Brussel, Brussels, Belgium
| | - Joeri L Aerts
- Laboratory of Molecular and Cellular Therapy; Department of Immunology-Physiology; Vrije Universiteit Brussel , Brussels, Belgium
| |
Collapse
|
200
|
Rumble JM, Huber AK, Krishnamoorthy G, Srinivasan A, Giles DA, Zhang X, Wang L, Segal BM. Neutrophil-related factors as biomarkers in EAE and MS. ACTA ACUST UNITED AC 2015; 212:23-35. [PMID: 25559893 PMCID: PMC4291533 DOI: 10.1084/jem.20141015] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Using a mouse model of multiple sclerosis (MS), the authors show that neutrophils expand in the bone marrow and accumulate in the circulation before clinical onset of disease. Early in disease development, neutrophils infiltrate the CNS, which is suppressed by G-CSF receptor deficiency and blockade of CXCL1 to ameliorate disease. In patients with MS, systemic expression of neutrophil-related mediators correlates with new lesion formation, lesion burden, and clinical disability. A major function of T helper (Th) 17 cells is to induce the production of factors that activate and mobilize neutrophils. Although Th17 cells have been implicated in the pathogenesis of multiple sclerosis (MS) and the animal model experimental autoimmune encephalomyelitis (EAE), little attention has been focused on the role of granulocytes in those disorders. We show that neutrophils, as well as monocytes, expand in the bone marrow and accumulate in the circulation before the clinical onset of EAE, in response to systemic up-regulation of granulocyte colony-stimulating factor (G-CSF) and the ELR+ CXC chemokine CXCL1. Neutrophils comprised a relatively high percentage of leukocytes infiltrating the central nervous system (CNS) early in disease development. G-CSF receptor deficiency and CXCL1 blockade suppressed myeloid cell accumulation in the blood and ameliorated the clinical course of mice that were injected with myelin-reactive Th17 cells. In relapsing MS patients, plasma levels of CXCL5, another ELR+ CXC chemokine, were elevated during acute lesion formation. Systemic expression of CXCL1, CXCL5, and neutrophil elastase correlated with measures of MS lesion burden and clinical disability. Based on these results, we advocate that neutrophil-related molecules be further investigated as novel biomarkers and therapeutic targets in MS.
Collapse
Affiliation(s)
- Julie M Rumble
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Amanda K Huber
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | | | - Ashok Srinivasan
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - David A Giles
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Xu Zhang
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Lu Wang
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Benjamin M Segal
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109 Holtom-Garrett Program in Neuroimmunology, Department of Neurology,Department of Radiology, Department of Biostatistics, and Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109 Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105
| |
Collapse
|