151
|
Sato R, Reuter T, Hiranuma R, Shibata T, Fukui R, Motoi Y, Murakami Y, Tsukamoto H, Yamazaki S, Liu K, Saitoh SI, Latz E, Miyake K. The impact of cell maturation and tissue microenvironments on the expression of endosomal Toll-like receptors in monocytes and macrophages. Int Immunol 2020; 32:785-798. [PMID: 32840578 DOI: 10.1093/intimm/dxaa055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 08/14/2020] [Indexed: 01/08/2023] Open
Abstract
Toll-like receptors (TLRs) impact myeloid cell responsiveness to environmental cues such as pathogen components and metabolites. Although TLR protein expression in monocytes and tissue macrophages is thought to be optimized for microenvironments in each tissue, a comprehensive study has not been reported. We here examined protein expression of endogenous TLRs in tissue-resident myeloid cells. Neutrophils in peripheral blood, spleen, liver and lung expressed TLR2, TLR4 and TLR5 in all tissues. Ly6C+ MHC II‒ classical monocytes mature into Ly6C‒ MHC II+ monocyte-derived dendritic cells (moDCs) or Ly6C‒ MHC II‒ patrolling monocytes. These subsets were found in all the tissues studied. TLR2 and TLR4 were displayed on all of these subsets, regardless of location. In contrast, expression of endosomal TLRs did vary with tissues and subsets. moDCs expressed TLR9, but much less TLR7. In contrast, TLR7, not TLR3 or TLR9, was highly expressed in classical and patrolling monocytes. Tissue macrophages such as red pulp macrophages in the spleen, Kupffer cells in the liver, microglia in the brain, alveolar macrophages in the lung and adipose tissue macrophages all expressed TLR2, TLR4 and TLR3. TLR7 was also expressed in these tissue macrophages except Kupffer cells in the liver. TLR9 expression in tissue macrophages was much lower or hard to detect. These results suggest that expression of endosomal TLRs in myeloid cells is influenced by their differentiation status and tissue-specific microenvironments.
Collapse
Affiliation(s)
- Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Tatjana Reuter
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan.,Institute of Innate Immunity, Biomedical Center, Venusberg-Campus, University of Bonn, Bonn, Germany
| | - Ryosuke Hiranuma
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Takuma Shibata
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Yuji Motoi
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Yusuke Murakami
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Hiroki Tsukamoto
- Laboratory of Oncology, Pharmacy Practice and Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.,Department of Pharmaceutical Sciences, School of Pharmacy at Fukuoka, International University of Health and Welfare, Fukuoka, Japan
| | - Satoshi Yamazaki
- Laboratory of Stem Cell Therapy, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kaiwen Liu
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Shin-Ichiroh Saitoh
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Eicke Latz
- Institute of Innate Immunity, Biomedical Center, Venusberg-Campus, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Infectious Diseases and Immunology, UMass Medical School, Worcester, MA, USA
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, Japan
| |
Collapse
|
152
|
Porcuna J, Menéndez-Gutiérrez MP, Ricote M. Molecular control of tissue-resident macrophage identity by nuclear receptors. Curr Opin Pharmacol 2020; 53:27-34. [DOI: 10.1016/j.coph.2020.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/31/2022]
|
153
|
Alivernini S, MacDonald L, Elmesmari A, Finlay S, Tolusso B, Gigante MR, Petricca L, Di Mario C, Bui L, Perniola S, Attar M, Gessi M, Fedele AL, Chilaka S, Somma D, Sansom SN, Filer A, McSharry C, Millar NL, Kirschner K, Nerviani A, Lewis MJ, Pitzalis C, Clark AR, Ferraccioli G, Udalova I, Buckley CD, Gremese E, McInnes IB, Otto TD, Kurowska-Stolarska M. Distinct synovial tissue macrophage subsets regulate inflammation and remission in rheumatoid arthritis. Nat Med 2020; 26:1295-1306. [PMID: 32601335 DOI: 10.1038/s41591-020-0939-8] [Citation(s) in RCA: 392] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 05/12/2020] [Indexed: 12/28/2022]
Abstract
Immune-regulatory mechanisms of drug-free remission in rheumatoid arthritis (RA) are unknown. We hypothesized that synovial tissue macrophages (STM), which persist in remission, contribute to joint homeostasis. We used single-cell transcriptomics to profile 32,000 STMs and identified phenotypic changes in patients with early/active RA, treatment-refractory/active RA and RA in sustained remission. Each clinical state was characterized by different frequencies of nine discrete phenotypic clusters within four distinct STM subpopulations with diverse homeostatic, regulatory and inflammatory functions. This cellular atlas, combined with deep-phenotypic, spatial and functional analyses of synovial biopsy fluorescent activated cell sorted STMs, revealed two STM subpopulations (MerTKposTREM2high and MerTKposLYVE1pos) with unique remission transcriptomic signatures enriched in negative regulators of inflammation. These STMs were potent producers of inflammation-resolving lipid mediators and induced the repair response of synovial fibroblasts in vitro. A low proportion of MerTKpos STMs in remission was associated with increased risk of disease flare after treatment cessation. Therapeutic modulation of MerTKpos STM subpopulations could therefore be a potential treatment strategy for RA.
Collapse
MESH Headings
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Biopsy
- Cell Lineage/genetics
- Humans
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Joints/immunology
- Joints/metabolism
- Joints/pathology
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Macrophages/immunology
- Macrophages/metabolism
- Mannose Receptor
- Mannose-Binding Lectins/genetics
- Mannose-Binding Lectins/immunology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Synovial Fluid/immunology
- Synovial Fluid/metabolism
- Synovial Membrane
Collapse
Affiliation(s)
- Stefano Alivernini
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), .
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
- Institute of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy.
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK.
| | - Lucy MacDonald
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Aziza Elmesmari
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Samuel Finlay
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Barbara Tolusso
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Rita Gigante
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Petricca
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Clara Di Mario
- Institute of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Laura Bui
- Division of Pathology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Simone Perniola
- Institute of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Moustafa Attar
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Marco Gessi
- Division of Pathology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Laura Fedele
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Sabarinadh Chilaka
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Domenico Somma
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Stephen N Sansom
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Filer
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK
| | - Charles McSharry
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Neal L Millar
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | | | - Alessandra Nerviani
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Andrew R Clark
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | | | - Irina Udalova
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Christopher D Buckley
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK
| | - Elisa Gremese
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Institute of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Iain B McInnes
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE)
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Thomas D Otto
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), .
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK.
| | - Mariola Kurowska-Stolarska
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), .
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK.
| |
Collapse
|
154
|
Uehata T, Takeuchi O. RNA Recognition and Immunity-Innate Immune Sensing and Its Posttranscriptional Regulation Mechanisms. Cells 2020; 9:cells9071701. [PMID: 32708595 PMCID: PMC7407594 DOI: 10.3390/cells9071701] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
RNA acts as an immunostimulatory molecule in the innate immune system to activate nucleic acid sensors. It functions as an intermediate, conveying genetic information to control inflammatory responses. A key mechanism for RNA sensing is discriminating self from non-self nucleic acids to initiate antiviral responses reliably, including the expression of type I interferon (IFN) and IFN-stimulated genes. Another important aspect of the RNA-mediated inflammatory response is posttranscriptional regulation of gene expression, where RNA-binding proteins (RBPs) have essential roles in various RNA metabolisms, including splicing, nuclear export, modification, and translation and mRNA degradation. Recent evidence suggests that the control of mRNA stability is closely involved in signal transduction and orchestrates immune responses. In this study, we review the current understanding of how RNA is sensed by host RNA sensing machinery and discuss self/non-self-discrimination in innate immunity focusing on mammalian species. Finally, we discuss how posttranscriptional regulation by RBPs shape immune reactions.
Collapse
|
155
|
Jain N, Moeller J, Vogel V. Mechanobiology of Macrophages: How Physical Factors Coregulate Macrophage Plasticity and Phagocytosis. Annu Rev Biomed Eng 2020; 21:267-297. [PMID: 31167103 DOI: 10.1146/annurev-bioeng-062117-121224] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In addition to their early-recognized functions in host defense and the clearance of apoptotic cell debris, macrophages play vital roles in tissue development, homeostasis, and repair. If misregulated, they steer the progression of many inflammatory diseases. Much progress has been made in understanding the mechanisms underlying macrophage signaling, transcriptomics, and proteomics, under physiological and pathological conditions. Yet, the detailed mechanisms that tune circulating monocytes/macrophages and tissue-resident macrophage polarization, differentiation, specification, and their functional plasticity remain elusive. We review how physical factors affect macrophage phenotype and function, including how they hunt for particles and pathogens, as well as the implications for phagocytosis, autophagy, and polarization from proinflammatory to prohealing phenotype. We further discuss how this knowledge can be harnessed in regenerative medicine and for the design of new drugs and immune-modulatory drug delivery systems, biomaterials, and tissue scaffolds.
Collapse
Affiliation(s)
- Nikhil Jain
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, and Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland;
| | - Jens Moeller
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, and Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland;
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, and Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland;
| |
Collapse
|
156
|
Song P, Zhao Q, Zou MH. Targeting senescent cells to attenuate cardiovascular disease progression. Ageing Res Rev 2020; 60:101072. [PMID: 32298812 DOI: 10.1016/j.arr.2020.101072] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) is the most common disease to increase as life expectancy increases. Most high-profile pharmacological treatments for age-related CVD have led to inefficacious results, implying that novel approaches to treating these pathologies are needed. Emerging data have demonstrated that senescent cardiovascular cells, which are characterized by irreversible cell cycle arrest and a distinct senescence-associated secretory phenotype, accumulate in aged or diseased cardiovascular systems, suggesting that they may impair cardiovascular function. This review discusses the evidence implicating senescent cells in cardiovascular ageing, the onset and progression of CVD, and the molecular mechanisms underlying cardiovascular cell senescence. We also review eradication of senescent cardiovascular cells by small-molecule-drug-mediated apoptosis and immune cell-mediated efferocytosis and toxicity as promising and precisely targeted therapeutics for CVD prevention and treatment.
Collapse
|
157
|
Bain CC, Gibson DA, Steers NJ, Boufea K, Louwe PA, Doherty C, González-Huici V, Gentek R, Magalhaes-Pinto M, Shaw T, Bajénoff M, Bénézech C, Walmsley SR, Dockrell DH, Saunders PTK, Batada NN, Jenkins SJ. Rate of replenishment and microenvironment contribute to the sexually dimorphic phenotype and function of peritoneal macrophages. Sci Immunol 2020; 5:eabc4466. [PMID: 32561560 PMCID: PMC7610697 DOI: 10.1126/sciimmunol.abc4466] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
Macrophages reside in the body cavities where they maintain serosal homeostasis and provide immune surveillance. Peritoneal macrophages are implicated in the etiology of pathologies including peritonitis, endometriosis, and metastatic cancer; thus, understanding the factors that govern their behavior is vital. Using a combination of fate mapping techniques, we have investigated the impact of sex and age on murine peritoneal macrophage differentiation, turnover, and function. We demonstrate that the sexually dimorphic replenishment of peritoneal macrophages from the bone marrow, which is high in males and very low in females, is driven by changes in the local microenvironment that arise upon sexual maturation. Population and single-cell RNA sequencing revealed marked dimorphisms in gene expression between male and female peritoneal macrophages that was, in part, explained by differences in composition of these populations. By estimating the time of residency of different subsets within the cavity and assessing development of dimorphisms with age and in monocytopenic Ccr2 -/- mice, we demonstrate that key sex-dependent features of peritoneal macrophages are a function of the differential rate of replenishment from the bone marrow, whereas others are reliant on local microenvironment signals. We demonstrate that the dimorphic turnover of peritoneal macrophages contributes to differences in the ability to protect against pneumococcal peritonitis between the sexes. These data highlight the importance of considering both sex and age in susceptibility to inflammatory and infectious diseases.
Collapse
Affiliation(s)
- C C Bain
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK.
| | - D A Gibson
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - N J Steers
- Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - K Boufea
- Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - P A Louwe
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - C Doherty
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - V González-Huici
- Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - R Gentek
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, INSERM, U1104, CNRS UMR7280, 13288 Marseille, France
| | - M Magalhaes-Pinto
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
| | - T Shaw
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
- Manchester Collaborative Centre for Inflammation Research (MCCIR), University of Manchester, Manchester, UK
| | - M Bajénoff
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, INSERM, U1104, CNRS UMR7280, 13288 Marseille, France
| | - C Bénézech
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - S R Walmsley
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - D H Dockrell
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - P T K Saunders
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - N N Batada
- Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - S J Jenkins
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
158
|
Madenspacher JH, Morrell ED, Gowdy KM, McDonald JG, Thompson BM, Muse G, Martinez J, Thomas S, Mikacenic C, Nick JA, Abraham E, Garantziotis S, Stapleton RD, Meacham JM, Thomassen MJ, Janssen WJ, Cook DN, Wurfel MM, Fessler MB. Cholesterol 25-hydroxylase promotes efferocytosis and resolution of lung inflammation. JCI Insight 2020; 5:137189. [PMID: 32343675 DOI: 10.1172/jci.insight.137189] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Alveolar macrophages (AM) play a central role in initiation and resolution of lung inflammation, but the integration of these opposing core functions is poorly understood. AM expression of cholesterol 25-hydroxylase (CH25H), the primary biosynthetic enzyme for 25-hydroxycholesterol (25HC), far exceeds the expression of macrophages in other tissues, but no role for CH25H has been defined in lung biology. As 25HC is an agonist for the antiinflammatory nuclear receptor, liver X receptor (LXR), we speculated that CH25H might regulate inflammatory homeostasis in the lung. Here, we show that, of natural oxysterols or sterols, 25HC is induced in the inflamed lung of mice and humans. Ch25h-/- mice fail to induce 25HC and LXR target genes in the lung after LPS inhalation and exhibit delayed resolution of airway neutrophilia, which can be rescued by systemic treatment with either 25HC or synthetic LXR agonists. LXR-null mice also display delayed resolution, suggesting that native oxysterols promote resolution. During resolution, Ch25h is induced in macrophages upon their encounter with apoptotic cells and is required for LXR-dependent prevention of AM lipid overload, induction of Mertk, efferocytic resolution of airway neutrophilia, and induction of TGF-β. CH25H/25HC/LXR is, thus, an inducible metabolic axis that programs AMs for efferocytic resolution of inflammation.
Collapse
Affiliation(s)
- Jennifer H Madenspacher
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| | - Eric D Morrell
- Section of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, Seattle, Washington, USA
| | - Kymberly M Gowdy
- Division of Pulmonary, Critical Care and Sleep Medicine, and.,Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bonne M Thompson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ginger Muse
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| | - Jennifer Martinez
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| | - Seddon Thomas
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| | - Carmen Mikacenic
- Section of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, Seattle, Washington, USA
| | - Jerry A Nick
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Edward Abraham
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Stavros Garantziotis
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| | - Renee D Stapleton
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Julie M Meacham
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| | - Mary Jane Thomassen
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - William J Janssen
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Donald N Cook
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| | - Mark M Wurfel
- Section of Pulmonary, Critical Care, and Sleep Medicine, Harborview Medical Center, Seattle, Washington, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| |
Collapse
|
159
|
Tilstra JS, John S, Gordon RA, Leibler C, Kashgarian M, Bastacky S, Nickerson KM, Shlomchik MJ. B cell-intrinsic TLR9 expression is protective in murine lupus. J Clin Invest 2020; 130:3172-3187. [PMID: 32191633 PMCID: PMC7260024 DOI: 10.1172/jci132328] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 03/05/2020] [Indexed: 12/26/2022] Open
Abstract
Toll-like receptor 9 (TLR9) is a regulator of disease pathogenesis in systemic lupus erythematosus (SLE). Why TLR9 represses disease while TLR7 and MyD88 have the opposite effect remains undefined. To begin to address this question, we created 2 alleles to manipulate TLR9 expression, allowing for either selective deletion or overexpression. We used these to test cell type-specific effects of Tlr9 expression on the regulation of SLE pathogenesis. Notably, Tlr9 deficiency in B cells was sufficient to exacerbate nephritis while extinguishing anti-nucleosome antibodies, whereas Tlr9 deficiency in dendritic cells (DCs), plasmacytoid DCs, and neutrophils had no discernable effect on disease. Thus, B cell-specific Tlr9 deficiency unlinked disease from autoantibody production. Critically, B cell-specific Tlr9 overexpression resulted in ameliorated nephritis, opposite of the effect of deleting Tlr9. Our findings highlight the nonredundant role of B cell-expressed TLR9 in regulating lupus and suggest therapeutic potential in modulating and perhaps even enhancing TLR9 signals in B cells.
Collapse
Affiliation(s)
- Jeremy S. Tilstra
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shinu John
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Rachael A. Gordon
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Claire Leibler
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michael Kashgarian
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sheldon Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kevin M. Nickerson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mark J. Shlomchik
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
160
|
Characterization of splenic MRC1 hiMHCII lo and MRC1 loMHCII hi cells from the monocyte/macrophage lineage of White Leghorn chickens. Vet Res 2020; 51:73. [PMID: 32460863 PMCID: PMC7251834 DOI: 10.1186/s13567-020-00795-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 05/12/2020] [Indexed: 02/07/2023] Open
Abstract
Monocytes/macrophages, which are found in a variety of organs, maintain tissue homeostasis at a steady state and act as the first line of defence during pathogen-induced inflammation in the host. Most monocyte/macrophage lineage studies in chickens have been largely performed using cell lines, while few studies using primary cells have been conducted. In the present study, the phenotypic and functional characteristics of splenic monocyte/macrophage lineage cells during steady state and inflammatory conditions were examined. Splenic monocyte/macrophage lineage cells could be identified as MRC1loMHCIIhi and MRC1hiMHCIIlo cells based on their surface expression of MRC1 and MHCII. In the steady state, MRC1loMHCIIhi cells were more frequently found among MRC1+ cells. MRC1loMHCIIhi cells expressed a higher number of antigen-presenting molecules (MHCII, MHCI, and CD80) than MRC1hiMHCIIlo cells. In contrast, MRC1hiMHCIIlo cells showed better phagocytic and CCR5-dependent migratory properties than MRC1loMHCIIhi cells. Furthermore, MRC1hiMHCIIlo cells infiltrated the spleen in vivo and then became MRC1loMHCIIhi cells. During lipopolysaccharide (LPS)-induced inflammatory conditions that were produced via intraperitoneal (i.p.) injection, the proportion and absolute number of MRC1hiMHCIIlo cells were increased in the spleen. Uniquely, inflammation induced the downregulation of MHCII expression in MRC1hiMHCIIlo cells. The major source of inflammatory cytokines (IL-1β, IL-6, and IL-12) was MRC1loMHCIIhi cells. Furthermore, MRC1hiMHCIIlo cells showed greater bactericidal activity than MRC1loMHCIIhi cells during LPS-induced inflammation. Collectively, these results suggest that two subsets of monocyte/macrophage lineage cells exist in the chicken spleen that have functional differences.
Collapse
|
161
|
Cao X, Yakala GK, van den Hil FE, Cochrane A, Mummery CL, Orlova VV. Differentiation and Functional Comparison of Monocytes and Macrophages from hiPSCs with Peripheral Blood Derivatives. Stem Cell Reports 2020; 12:1282-1297. [PMID: 31189095 PMCID: PMC6565887 DOI: 10.1016/j.stemcr.2019.05.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/06/2019] [Accepted: 05/06/2019] [Indexed: 12/19/2022] Open
Abstract
A renewable source of human monocytes and macrophages would be a valuable alternative to primary cells from peripheral blood (PB) in biomedical research. We developed an efficient protocol to derive monocytes and macrophages from human induced pluripotent stem cells (hiPSCs) and performed a functional comparison with PB-derived cells. hiPSC-derived monocytes were functional after cryopreservation and exhibited gene expression profiles comparable with PB-derived monocytes. Notably, hiPSC-derived monocytes were more activated with greater adhesion to endothelial cells under physiological flow. hiPSC-derived monocytes were successfully polarized to M1 and M2 macrophage subtypes, which showed similar pan- and subtype-specific gene and surface protein expression and cytokine secretion to PB-derived macrophages. hiPSC-derived macrophages exhibited higher endocytosis and efferocytosis and similar bacterial and tumor cell phagocytosis to PB-derived macrophages. In summary, we developed a robust protocol to generate hiPSC monocytes and macrophages from independent hiPSC lines that showed aspects of functional maturity comparable with those from PB.
Collapse
Affiliation(s)
- Xu Cao
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Gopala K Yakala
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Amy Cochrane
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands.
| |
Collapse
|
162
|
Hine AM, Loke P. Intestinal Macrophages in Resolving Inflammation. THE JOURNAL OF IMMUNOLOGY 2020; 203:593-599. [PMID: 31332080 DOI: 10.4049/jimmunol.1900345] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/22/2019] [Indexed: 12/15/2022]
Abstract
Macrophages not only regulate intestinal homeostasis by recognizing pathogens to control enteric infections but also employ negative feedback mechanisms to prevent chronic inflammation. Hence, macrophages are intriguing targets for immune-mediated therapies, especially when barrier function in the gut is compromised to trigger aberrant inflammatory responses, most notably during inflammatory bowel diseases. Recently, there has been considerable progress in our understanding of human macrophage biology in different tissues, including the intestines. In this review, we discuss some new findings on the properties of distinct populations of intestinal macrophages, how resolution of inflammation and tissue repair by macrophages could be promoted by type 2 cytokines as well as other therapeutic interventions, and highlight some challenges for translating these findings into the future for this exciting area of immunology research.
Collapse
Affiliation(s)
- Ashley M Hine
- Department of Microbiology, New York University School of Medicine, New York, NY 10016
| | - P'ng Loke
- Department of Microbiology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
163
|
PAM3 protects against DSS-induced colitis by altering the M2:M1 ratio. Sci Rep 2020; 10:6078. [PMID: 32269253 PMCID: PMC7142137 DOI: 10.1038/s41598-020-63143-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/16/2020] [Indexed: 12/26/2022] Open
Abstract
Inflammation of the gastrointestinal tract contributes to the development of inflammatory bowel disease (IBD). Human IBD is modeled by administering dextran sulfate sodium (DSS) to mice. In humans and mice, inflammatory M1 macrophages contribute to the progression of IBD whereas immunosuppressive M2 macrophages protect against colitis. The TLR2/1 agonist PAM3CSK4 (PAM3) induces human and murine monocytes to differentiate into immunosuppressive M2 macrophages, suggesting that PAM3 might be of benefit in the prevention/treatment of colitis. PAM3 was therefore administered to mice treated with DSS. As hypothesized, the number of M2 macrophages rose and disease severity decreased. The critical role of M2 macrophages in this process was established by transferring purified M2 macrophages from PAM3 treated control donors into DSS recipients and reducing colitis. These findings suggest that PAM3 may represent a novel approach to the treatment of human IBD.
Collapse
|
164
|
RXRs control serous macrophage neonatal expansion and identity and contribute to ovarian cancer progression. Nat Commun 2020; 11:1655. [PMID: 32246014 PMCID: PMC7125161 DOI: 10.1038/s41467-020-15371-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/28/2020] [Indexed: 12/04/2022] Open
Abstract
Tissue-resident macrophages (TRMs) populate all tissues and play key roles in homeostasis, immunity and repair. TRMs express a molecular program that is mostly shaped by tissue cues. However, TRM identity and the mechanisms that maintain TRMs in tissues remain poorly understood. We recently found that serous-cavity TRMs (LPMs) are highly enriched in RXR transcripts and RXR-response elements. Here, we show that RXRs control mouse serous-macrophage identity by regulating chromatin accessibility and the transcriptional regulation of canonical macrophage genes. RXR deficiency impairs neonatal expansion of the LPM pool and reduces the survival of adult LPMs through excess lipid accumulation. We also find that peritoneal LPMs infiltrate early ovarian tumours and that RXR deletion diminishes LPM accumulation in tumours and strongly reduces ovarian tumour progression in mice. Our study reveals that RXR signalling controls the maintenance of the serous macrophage pool and that targeting peritoneal LPMs may improve ovarian cancer outcomes. Macrophages can differentiate to perform homeostatic tissue-specific functions. Here the authors show that RXR signalling is critical for large peritoneal macrophage (LPM) expansion during neonatal life and LPM lipid metabolism and survival during adult homeostasis, and that ovarian cancer growth relies on RXR-dependent LPMs.
Collapse
|
165
|
Inokuchi S, Mitoma H, Kawano S, Nakano S, Ayano M, Kimoto Y, Akahoshi M, Arinobu Y, Tsukamoto H, Akashi K, Horiuchi T, Niiro H. Homeostatic Milieu Induces Production of Deoxyribonuclease 1–like 3 from Myeloid Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:2088-2097. [DOI: 10.4049/jimmunol.1901304] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/12/2020] [Indexed: 12/31/2022]
|
166
|
Immune Clearance of Senescent Cells to Combat Ageing and Chronic Diseases. Cells 2020; 9:cells9030671. [PMID: 32164335 PMCID: PMC7140645 DOI: 10.3390/cells9030671] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/28/2020] [Accepted: 03/05/2020] [Indexed: 12/19/2022] Open
Abstract
Senescent cells are generally characterized by permanent cell cycle arrest, metabolic alteration and activation, and apoptotic resistance in multiple organs due to various stressors. Excessive accumulation of senescent cells in numerous tissues leads to multiple chronic diseases, tissue dysfunction, age-related diseases and organ ageing. Immune cells can remove senescent cells. Immunaging or impaired innate and adaptive immune responses by senescent cells result in persistent accumulation of various senescent cells. Although senolytics-drugs that selectively remove senescent cells by inducing their apoptosis-are recent hot topics and are making significant research progress, senescence immunotherapies using immune cell-mediated clearance of senescent cells are emerging and promising strategies to fight ageing and multiple chronic diseases. This short review provides an overview of the research progress to date concerning senescent cell-caused chronic diseases and tissue ageing, as well as the regulation of senescence by small-molecule drugs in clinical trials and different roles and regulation of immune cells in the elimination of senescent cells. Mounting evidence indicates that immunotherapy targeting senescent cells combats ageing and chronic diseases and subsequently extends the healthy lifespan.
Collapse
|
167
|
Chitu V, Biundo F, Shlager GGL, Park ES, Wang P, Gulinello ME, Gokhan Ş, Ketchum HC, Saha K, DeTure MA, Dickson DW, Wszolek ZK, Zheng D, Croxford AL, Becher B, Sun D, Mehler MF, Stanley ER. Microglial Homeostasis Requires Balanced CSF-1/CSF-2 Receptor Signaling. Cell Rep 2020; 30:3004-3019.e5. [PMID: 32130903 PMCID: PMC7370656 DOI: 10.1016/j.celrep.2020.02.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 12/18/2019] [Accepted: 02/06/2020] [Indexed: 02/08/2023] Open
Abstract
CSF-1R haploinsufficiency causes adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). Previous studies in the Csf1r+/- mouse model of ALSP hypothesized a central role of elevated cerebral Csf2 expression. Here, we show that monoallelic deletion of Csf2 rescues most behavioral deficits and histopathological changes in Csf1r+/- mice by preventing microgliosis and eliminating most microglial transcriptomic alterations, including those indicative of oxidative stress and demyelination. We also show elevation of Csf2 transcripts and of several CSF-2 downstream targets in the brains of ALSP patients, demonstrating that the mechanisms identified in the mouse model are functional in humans. Our data provide insights into the mechanisms underlying ALSP. Because increased CSF2 levels and decreased microglial Csf1r expression have also been reported in Alzheimer's disease and multiple sclerosis, we suggest that the unbalanced CSF-1R/CSF-2 signaling we describe in the present study may contribute to the pathogenesis of other neurodegenerative conditions.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Gabriel G L Shlager
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Eun S Park
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maria E Gulinello
- Behavioral Core Facility, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Şölen Gokhan
- Institute for Brain Disorders and Neural Regeneration, Departments of Neurology, Neuroscience, and Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Harmony C Ketchum
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kusumika Saha
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Michael A DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Deyou Zheng
- The Saul R. Korey Department of Neurology, Dominick P. Purpura Department of Neuroscience, and Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich 8057, Switzerland
| | - Daqian Sun
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Mark F Mehler
- Institute for Brain Disorders and Neural Regeneration, Departments of Neurology, Neuroscience, and Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
168
|
Tajbakhsh A, Rezaee M, Barreto GE, Moallem SA, Henney NC, Sahebkar A. The role of nuclear factors as “Find-Me”/alarmin signals and immunostimulation in defective efferocytosis and related disorders. Int Immunopharmacol 2020; 80:106134. [DOI: 10.1016/j.intimp.2019.106134] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/22/2022]
|
169
|
Dysregulation of TLR9 in neonates leads to fatal inflammatory disease driven by IFN-γ. Proc Natl Acad Sci U S A 2020; 117:3074-3082. [PMID: 31980536 DOI: 10.1073/pnas.1911579117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Recognition of self-nucleic acids by innate immune receptors can lead to the development of autoimmune and/or autoinflammatory diseases. Elucidating mechanisms associated with dysregulated activation of specific receptors may identify new disease correlates and enable more effective therapies. Here we describe an aggressive in vivo model of Toll-like receptor (TLR) 9 dysregulation, based on bypassing the compartmentalized activation of TLR9 in endosomes, and use it to uncover unique aspects of TLR9-driven disease. By inducing TLR9 dysregulation at different stages of life, we show that while dysregulation in adult mice causes a mild systemic autoinflammatory disease, dysregulation of TLR9 early in life drives a severe inflammatory disease resulting in neonatal fatality. The neonatal disease includes some hallmarks of macrophage activation syndrome but is much more severe than previously described models. Unlike TLR7-mediated disease, which requires type I interferon (IFN) receptor signaling, TLR9-driven fatality is dependent on IFN-γ receptor signaling. NK cells are likely key sources of IFN-γ in this model. We identify populations of macrophages and Ly6Chi monocytes in neonates that express high levels of TLR9 and low levels of TLR7, which may explain why TLR9 dysregulation is particularly consequential early in life, while symptoms of TLR7 dysregulation take longer to manifest. Overall, this study demonstrates that inappropriate TLR9 responses can drive a severe autoinflammatory disease under homeostatic conditions and highlights differences in the diseases resulting from inappropriate activation of TLR9 and TLR7.
Collapse
|
170
|
Blériot C, Ng LG, Ginhoux F. "Cloaking" on Time: A Cover-Up Act by Resident Tissue Macrophages. Cell 2020; 177:514-516. [PMID: 31002790 DOI: 10.1016/j.cell.2019.03.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In this issue of Cell, Uderhardt et al. employed intravital two-photon microscopy to examine tissue-resident macrophage responses to sterile cellular injuries of variable size. They observed that while multi-cell "macrolesions" are characteristically pro-inflammatory, resident macrophages can "cloak" single-cell microlesions to prevent excessive neutrophil recruitment and limit subsequent tissue damage.
Collapse
Affiliation(s)
- Camille Blériot
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), 8A Biomedical Grove, Immunos Building #3-4, Biopolis, Singapore 138648, Singapore
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), 8A Biomedical Grove, Immunos Building #3-4, Biopolis, Singapore 138648, Singapore; State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences, 288 Nanjing Road, Tianjin 300020, China
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), 8A Biomedical Grove, Immunos Building #3-4, Biopolis, Singapore 138648, Singapore; Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
171
|
Park JG, Lee CR, Kim MG, Kim G, Shin HM, Jeon YH, Yang SH, Kim DK, Joo KW, Choi EY, Kim HR, Kwak C, Kim YS, Choi M, Lee DS, Han SS. Kidney residency of VISTA-positive macrophages accelerates repair from ischemic injury. Kidney Int 2019; 97:980-994. [PMID: 32143848 DOI: 10.1016/j.kint.2019.11.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 11/21/2019] [Accepted: 11/27/2019] [Indexed: 01/04/2023]
Abstract
Tissue-resident macrophages have unique tissue-specific functions in maintaining homeostasis and resolving inflammation. However, the repair role and relevant molecules of kidney-resident macrophages after ischemic injury remain unresolved. To this end, mice without kidney-resident R1 macrophages but containing infiltrating monocyte-derived R2 macrophages were generated using differential cellular kinetics following clodronate liposome treatment. When ischemia-reperfusion injury was induced in these mice, late phase repair was reduced. Transcriptomic and flow cytometric analyses identified that V-domain Ig suppressor of T cell activation (VISTA), an inhibitory immune checkpoint molecule, was constitutively expressed in kidney-resident R1 macrophages, but not in other tissue-resident macrophages. Here, VISTA functioned as a scavenger of apoptotic cells and served as a checkpoint to control kidney-infiltrating T cells upon T cell receptor-mediated stimulation. Together these functions improved the repair process after ischemia-reperfusion injury. CD14+ CD33+ mononuclear phagocytes of human kidney also expressed VISTA, which has similar functions to the mouse counterpart. Thus, VISTA is upregulated in kidney macrophages in a tissue-dependent manner and plays a repair role during ischemic injury.
Collapse
Affiliation(s)
- Jun-Gyu Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Cho-Rong Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Min-Gang Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Gwanghun Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Mu Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Yun-Hui Jeon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Hee Yang
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Dong Ki Kim
- Kidney Research Institute, Seoul National University, Seoul, Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Hang-Rae Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Cheol Kwak
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea
| | - Yon Su Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea; Kidney Research Institute, Seoul National University, Seoul, Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea; Kidney Research Institute, Seoul National University, Seoul, Korea.
| | - Seung Seok Han
- Kidney Research Institute, Seoul National University, Seoul, Korea; Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
172
|
Nackiewicz D, Dan M, Speck M, Chow SZ, Chen YC, Pospisilik JA, Verchere CB, Ehses JA. Islet Macrophages Shift to a Reparative State following Pancreatic Beta-Cell Death and Are a Major Source of Islet Insulin-like Growth Factor-1. iScience 2019; 23:100775. [PMID: 31962237 PMCID: PMC6971395 DOI: 10.1016/j.isci.2019.100775] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 09/24/2019] [Accepted: 12/10/2019] [Indexed: 12/21/2022] Open
Abstract
Macrophages play a dynamic role in tissue repair following injury. Here we found that following streptozotocin (STZ)-induced beta-cell death, mouse islet macrophages had increased Igf1 expression, decreased proinflammatory cytokine expression, and transcriptome changes consistent with macrophages undergoing efferocytosis and having an enhanced state of metabolism. Macrophages were the major, if not sole, contributors to islet insulin-like growth factor-1 (IGF-1) production. Adoptive transfer experiments showed that macrophages can maintain insulin secretion in vivo following beta-cell death with no effects on islet cell turnover. IGF-1 neutralization during STZ treatment decreased insulin secretion without affecting islet cell apoptosis or proliferation. Interestingly, high-fat diet (HFD) combined with STZ further skewed islet macrophages to a reparative state. Finally, islet macrophages from db/db mice also expressed decreased proinflammatory cytokines and increased Igf1 mRNA. These data have important implications for islet biology and pathology and show that islet macrophages preserve their reparative state following beta-cell death even during HFD feeding and severe hyperglycemia. Macrophages are a major source of IGF-1 protein within mouse pancreatic islets Post-beta-cell death islet macrophages shift to a reparative state Beta-cell death causes macrophage transcriptome changes consistent with efferocytosis This change can occur even in the presence of HFD feeding or severe hyperglycemia
Collapse
Affiliation(s)
- Dominika Nackiewicz
- Department of Surgery, Faculty of Medicine, University of British Columbia, BC Children's Hospital Research Institute, 950 W 28 Avenue, Vancouver V5Z 4H4, Canada
| | - Meixia Dan
- Department of Surgery, Faculty of Medicine, University of British Columbia, BC Children's Hospital Research Institute, 950 W 28 Avenue, Vancouver V5Z 4H4, Canada
| | - Madeleine Speck
- Department of Surgery, Faculty of Medicine, University of British Columbia, BC Children's Hospital Research Institute, 950 W 28 Avenue, Vancouver V5Z 4H4, Canada
| | - Samuel Z Chow
- Department of Surgery, Faculty of Medicine, University of British Columbia, BC Children's Hospital Research Institute, 950 W 28 Avenue, Vancouver V5Z 4H4, Canada
| | - Yi-Chun Chen
- Department of Surgery, Faculty of Medicine, University of British Columbia, BC Children's Hospital Research Institute, 950 W 28 Avenue, Vancouver V5Z 4H4, Canada
| | - J Andrew Pospisilik
- Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, MI 49503, USA
| | - C Bruce Verchere
- Department of Surgery, Faculty of Medicine, University of British Columbia, BC Children's Hospital Research Institute, 950 W 28 Avenue, Vancouver V5Z 4H4, Canada; Department of Pathology and Laboratory Medicine, BC Children's Hospital Research Institute, 950 W 28 Avenue, Vancouver V5Z 4H4, Canada.
| | - Jan A Ehses
- Department of Surgery, Faculty of Medicine, University of British Columbia, BC Children's Hospital Research Institute, 950 W 28 Avenue, Vancouver V5Z 4H4, Canada; Department of Health Sciences and Technology, Institute of Food, Nutrition, and Health, Swiss Federal Institute of Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland.
| |
Collapse
|
173
|
van der Heide D, Weiskirchen R, Bansal R. Therapeutic Targeting of Hepatic Macrophages for the Treatment of Liver Diseases. Front Immunol 2019; 10:2852. [PMID: 31849997 PMCID: PMC6901832 DOI: 10.3389/fimmu.2019.02852] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022] Open
Abstract
Hepatic macrophages play a central role in maintaining homeostasis in the liver, as well as in the initiation and progression of liver diseases. Hepatic macrophages are mainly derived from resident hepatic macrophages called Kupffer cells or circulating bone marrow-derived monocytes. Kupffer cells are self-renewing and typically non-migrating macrophages in the liver and are stationed in the liver sinusoids in contrast to macrophages originating from circulating monocytes. Kupffer cells regulate liver homeostasis by mediating immunity against non-pathogenic blood-borne molecules, while participating in coordinated immune responses leading to pathogen clearance, leukocyte recruitment and antigen presentation to lymphocytes present in the vasculature. Monocyte-derived macrophages infiltrate into the liver tissue when metabolic or toxic damage instigates and are likely dispensable for replenishing the macrophage population in homeostasis. In recent years, different populations of hepatic macrophages have been identified with distinct phenotypes with discrete functions, far beyond the central dogma of M1 and M2 macrophages. Hepatic macrophages play a central role in the pathogenesis of acute and chronic liver failure, liver fibrosis, non-alcoholic fatty liver disease, alcoholic liver disease, viral hepatitis, and hepatocellular carcinoma, as well as in disease resolution. The understanding of the role of hepatic macrophages in liver diseases provides opportunities for the development of targeted therapeutics for respective malignancies. This review will summarize the current knowledge of the hepatic macrophages, their origin, functions, their critical role in maintaining homeostasis and in the progression or resolution of liver diseases. Furthermore, we will provide a comprehensive overview of the therapeutic targeting strategies against hepatic macrophages developed for the treatment of liver diseases.
Collapse
Affiliation(s)
- Daphne van der Heide
- Department of Biomaterials Science and Technology, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Ruchi Bansal
- Department of Biomaterials Science and Technology, Faculty of Science and Technology, Technical Medical Center, University of Twente, Enschede, Netherlands
| |
Collapse
|
174
|
Gyoneva S, Hosur R, Gosselin D, Zhang B, Ouyang Z, Cotleur AC, Peterson M, Allaire N, Challa R, Cullen P, Roberts C, Miao K, Reynolds TL, Glass CK, Burkly L, Ransohoff RM. Cx3cr1-deficient microglia exhibit a premature aging transcriptome. Life Sci Alliance 2019; 2:2/6/e201900453. [PMID: 31792059 PMCID: PMC6892408 DOI: 10.26508/lsa.201900453] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 11/14/2019] [Accepted: 11/17/2019] [Indexed: 12/13/2022] Open
Abstract
Gyoneva et al use RNA-seq to show that Cx3cr1-deficient microglia in young mice display a gene expression profile similar to microglia in aged mice, suggesting premature microglial aging. CX3CR1, one of the highest expressed genes in microglia in mice and humans, is implicated in numerous microglial functions. However, the molecular mechanisms underlying Cx3cr1 signaling are not well understood. Here, we analyzed transcriptomes of Cx3cr1-deficient microglia under varying conditions by RNA-sequencing (RNA-seq). In 2-mo-old mice, Cx3cr1 deletion resulted in the down-regulation of a subset of immune-related genes, without substantial epigenetic changes in markers of active chromatin. Surprisingly, Cx3cr1-deficient microglia from young mice exhibited a transcriptome consistent with that of aged Cx3cr1-sufficient animals, suggesting a premature aging transcriptomic signature. Immunohistochemical analysis of microglia in young and aged mice revealed that loss of Cx3cr1 modulates microglial morphology in a comparable fashion. Our results suggest that CX3CR1 may regulate microglial function in part by modulating the expression levels of a subset of inflammatory genes during chronological aging, making Cx3cr1-deficient mice useful for studying aged microglia.
Collapse
Affiliation(s)
| | | | - David Gosselin
- Cell and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | - Baohong Zhang
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Zhengyu Ouyang
- Cell and Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | | | | | - Norm Allaire
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Ravi Challa
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Patrick Cullen
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Chris Roberts
- Computational Biology and Genomics, Biogen, Cambridge, MA, USA
| | - Kelly Miao
- Acute Neurology, Biogen, Cambridge, MA, USA
| | | | - Christopher K Glass
- Cell and Molecular Medicine, University of California San Diego, San Diego, CA, USA.,School of Medicine, University of California San Diego, San Diego, CA, USA
| | | | | |
Collapse
|
175
|
Yang D, Xing Y, Song X, Qian Y. The impact of lung microbiota dysbiosis on inflammation. Immunology 2019; 159:156-166. [PMID: 31631335 DOI: 10.1111/imm.13139] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/05/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Host-microbiota interaction plays fundamental roles in the homeostasis of mucosal immunity. Dysbiosis of intestinal microbiota has been demonstrated to participate in various immune responses and many multifactorial diseases. Study of intestinal microbiota has moved beyond the consequences of dysbiosis to the causal microbiota associated with diseases. However, studies of pulmonary microbiota and its dysbiosis are still in their infancy. Improvement of culture-dependent and -independent techniques has facilitated our understanding of lung microbiota that not only exists in healthy lung tissue but also exerts great impact on immune responses under both physiological and pathological conditions. In this review, we summarize recent progresses of lung microbiota dysbiosis and its impact on the local immune system that determines the balance of tolerance and inflammation. We discuss the causal roles of pulmonary dysbiosis under disease settings, and propose that the interaction between lung microbiota and host is critical for establishing the immune homeostasis in lung.
Collapse
Affiliation(s)
- Daping Yang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yingying Xing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xinyang Song
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Youcun Qian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
176
|
Plazyo O, Sheng JJ, Jin JP. Downregulation of calponin 2 contributes to the quiescence of lung macrophages. Am J Physiol Cell Physiol 2019; 317:C749-C761. [PMID: 31365293 PMCID: PMC6850996 DOI: 10.1152/ajpcell.00036.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/08/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022]
Abstract
Calponin 2 is an actin cytoskeleton-associated regulatory protein that inhibits the activity of myosin-ATPase and cytoskeleton dynamics. Recent studies have demonstrated that deletion of calponin 2 restricts the proinflammatory activation of macrophages in atherosclerosis and arthritis to attenuate the disease progression in mice. Here we demonstrate that the levels of calponin 2 vary among different macrophage populations, which may reflect their adaptation to specific tissue microenvironment corresponding to specific functional states. Interestingly, lung resident macrophages express significantly lower calponin 2 than peritoneal resident macrophages, which correlates with decreased substrate adhesion and reduced expression of proinflammatory cytokines and a proresolution phenotype. Deletion of calponin 2 in peritoneal macrophages also decreased substrate adhesion and downregulated the expression of proinflammatory cytokines. Providing the first line of defense against microbial invasion while receiving constant exposure to extrinsic antigens, lung macrophages need to maintain a necessary level of activity while limiting exaggerated inflammatory reaction. Therefore, their low level of calponin 2 may reflect an important physiological adaption. Downregulation of calponin 2 in macrophages may be targeted as a cytoskeleton-based novel mechanism, possibly via endoplasmic reticulum stress altering the processing and secretion of cytokines, to regulate immune response and promote quiescence for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Olesya Plazyo
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - Juan-Juan Sheng
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| | - J-P Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
177
|
Ikeda N, Asano K, Kikuchi K, Uchida Y, Ikegami H, Takagi R, Yotsumoto S, Shibuya T, Makino-Okamura C, Fukuyama H, Watanabe T, Ohmuraya M, Araki K, Nishitai G, Tanaka M. Emergence of immunoregulatory Ym1 +Ly6C hi monocytes during recovery phase of tissue injury. Sci Immunol 2019; 3:3/28/eaat0207. [PMID: 30291130 DOI: 10.1126/sciimmunol.aat0207] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/25/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022]
Abstract
Ly6Chi monocytes migrate to injured sites and induce inflammation in the acute phase of tissue injury. However, once the causes of tissue injury are eliminated, monocyte-derived macrophages contribute to the resolution of inflammation and tissue repair. It remains unclear whether the emergence of these immunoregulatory macrophages is attributed to the phenotypic conversion of inflammatory monocytes in situ or to the recruitment of bone marrow-derived regulatory cells de novo. Here, we identified a subpopulation of Ly6Chi monocytes that contribute to the resolution of inflammation and tissue repair. Ym1+Ly6Chi monocytes greatly expanded in bone marrow during the recovery phase of systemic inflammation or tissue injury. Ym1+Ly6Chi monocytes infiltrating into an injured site exhibited immunoregulatory and tissue-reparative phenotypes. Deletion of Ym1+Ly6Chi monocytes resulted in delayed recovery from colitis. These results demonstrate that a distinct monocyte subpopulation destined to act in immunoregulation is generated in bone marrow and participates in resolution of inflammation and tissue repair.
Collapse
Affiliation(s)
- Naoki Ikeda
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Kenichi Asano
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Kenta Kikuchi
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Yoshimi Uchida
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Hiroki Ikegami
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Ryo Takagi
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Satoshi Yotsumoto
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Takumi Shibuya
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Chieko Makino-Okamura
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Hidehiro Fukuyama
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Takashi Watanabe
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Masaki Ohmuraya
- Department of Genetics, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Kimi Araki
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Kumamoto, 860-0811, Japan
| | - Gen Nishitai
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Masato Tanaka
- Laboratory of Immune Regulation, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan.
| |
Collapse
|
178
|
UNC93B1 recruits syntenin-1 to dampen TLR7 signalling and prevent autoimmunity. Nature 2019; 575:366-370. [PMID: 31546246 PMCID: PMC6856441 DOI: 10.1038/s41586-019-1612-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/23/2019] [Indexed: 12/21/2022]
Abstract
At least two members of the Toll-like receptor (TLR) family, TLR7 and TLR9, can recognize self-RNA and self-DNA, respectively. Despite the structural and functional similarities between these receptors, their contributions to autoimmune diseases such as systemic lupus erythematosus can differ. For example, TLR7 and TLR9 have opposing effects in mouse models of systemic lupus erythematosus-disease is exacerbated in TLR9-deficient mice but attenuated in TLR7-deficient mice1. However, the mechanisms of negative regulation that differentiate between TLR7 and TLR9 are unknown. Here we report a function for the TLR trafficking chaperone UNC93B1 that specifically limits signalling of TLR7, but not TLR9, and prevents TLR7-dependent autoimmunity in mice. Mutations in UNC93B1 that lead to enhanced TLR7 signalling also disrupt binding of UNC93B1 to syntenin-1, which has been implicated in the biogenesis of exosomes2. Both UNC93B1 and TLR7 can be detected in exosomes, suggesting that recruitment of syntenin-1 by UNC93B1 facilitates the sorting of TLR7 into intralumenal vesicles of multivesicular bodies, which terminates signalling. Binding of syntenin-1 requires phosphorylation of UNC93B1 and provides a mechanism for dynamic regulation of TLR7 activation and signalling. Thus, UNC93B1 not only enables the proper trafficking of nucleic acid-sensing TLRs, but also sets the activation threshold of potentially self-reactive TLR7.
Collapse
|
179
|
Release from UNC93B1 reinforces the compartmentalized activation of select TLRs. Nature 2019; 575:371-374. [PMID: 31546247 PMCID: PMC6856438 DOI: 10.1038/s41586-019-1611-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/12/2019] [Indexed: 11/24/2022]
Abstract
Nucleic acid-sensing Toll-like receptors (TLRs) are subject to complex regulation to facilitate recognition of microbial DNA and RNA while limiting recognition of self-nucleic acids1. Failure to properly regulate these TLRs can lead to autoimmune and autoinflammatory disease2–6. Intracellular localization of these receptors is thought to be critical for self vs. non-self discrimination7, yet the molecular mechanisms that reinforce compartmentalized activation of intracellular TLRs remain poorly understood. Here we describe a new mechanism that prevents TLR9 activation from locations other than endosomes. This control is achieved through the regulated release of the receptor from its trafficking chaperone Unc93b1, which only occurs within endosomes and is required for ligand binding and signal transduction. Preventing TLR9 release from Unc93b1, either through mutations in Unc93b1 that increase affinity for TLR9 or through an artificial tether that impairs release, results in defective signaling. While TLR9 and TLR3 release from Unc93b1, TLR7 does not dissociate from Unc93b1 in endosomes and is regulated via distinct mechanisms. This work defines a new checkpoint that reinforces compartmentalized activation of TLR9 and provides a mechanism by which activation of individual endosomal TLRs may be distinctly regulated.
Collapse
|
180
|
Culemann S, Grüneboom A, Krönke G. Origin and function of synovial macrophage subsets during inflammatory joint disease. Adv Immunol 2019; 143:75-98. [PMID: 31607368 DOI: 10.1016/bs.ai.2019.08.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mononuclear phagocytes, including monocytes and macrophages, are a central component of the host's innate immune system designated to protect against invading pathogens. However, these cells do not only interact with various parts of the innate and adaptive immune system, but also fulfill indispensable duties during the control of tissue homeostasis and organ function. Moreover, macrophages are crucially involved in tissue remodeling and repair in response to damage. Simultaneously, mononuclear phagocytes might also contribute to the pathogenesis of various inflammatory and autoimmune diseases. In particular, their potential role in inflammatory joint diseases such as rheumatoid arthritis (RA) has drawn increasing attention and substantially shaped our general understanding of the role of monocytes and macrophages during health and disease. This review summarizes our current knowledge about the origin and function of mononuclear phagocytes within the joint and addresses their involvement in joint inflammation.
Collapse
Affiliation(s)
- Stephan Culemann
- Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.; Nikolaus Fiebiger Center of Molecular Medicine, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anika Grüneboom
- Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.; Nikolaus Fiebiger Center of Molecular Medicine, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3-Rheumatology and Immunology, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.; Nikolaus Fiebiger Center of Molecular Medicine, Universitätsklinikum Erlangen and Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
181
|
Borst K, Graalmann T, Kalinke U. Reply to: "Unveiling the depletion of Kupffer cells in experimental hepatocarcinogenesis through liver macrophage subtype-specific markers". J Hepatol 2019; 71:633-635. [PMID: 31227257 DOI: 10.1016/j.jhep.2019.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/18/2019] [Accepted: 05/22/2019] [Indexed: 12/04/2022]
Affiliation(s)
- Katharina Borst
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Germany
| | - Theresa Graalmann
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Brunswick, Germany; Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Brunswick, Germany; Cluster of Excellence - Resolving Infection Susceptibility (RESIST), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
182
|
Kreuk LSM, Koch MA, Slayden LC, Lind NA, Chu S, Savage HP, Kantor AB, Baumgarth N, Barton GM. B cell receptor and Toll-like receptor signaling coordinate to control distinct B-1 responses to both self and the microbiota. eLife 2019; 8:e47015. [PMID: 31433298 PMCID: PMC6703855 DOI: 10.7554/elife.47015] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/16/2019] [Indexed: 01/19/2023] Open
Abstract
B-1a cells play an important role in mediating tissue homeostasis and protecting against infections. They are the main producers of 'natural' IgM, spontaneously secreted serum antibodies predominately reactive to self antigens, like phosphatidylcholine (PtC), or antigens expressed by the intestinal microbiota. The mechanisms that regulate the B-1a immunoglobulin (Ig) repertoire and their antibody secretion remain poorly understood. Here, we use a novel reporter mouse to demonstrate that production of self- and microbiota-reactive antibodies is linked to BCR signaling in B-1a cells. Moreover, we show that Toll-like receptors (TLRs) are critical for shaping the Ig repertoire of B-1a cells as well as regulating their antibody production. Strikingly, we find that both the colonization of a microbiota as well as microbial-sensing TLRs are required for anti-microbiota B-1a responses, whereas nucleic-acid sensing TLRs are required for anti-PtC responses, demonstrating that linked activation of BCR and TLRs controls steady state B-1a responses to both self and microbiota-derived antigens.
Collapse
Affiliation(s)
- Lieselotte SM Kreuk
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Meghan A Koch
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Leianna C Slayden
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Nicholas A Lind
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Sophia Chu
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| | - Hannah P Savage
- Center for Comparative MedicineUniversity of California, DavisDavisUnited States
| | - Aaron B Kantor
- Department of GeneticsStanford UniversityStanfordUnited States
| | - Nicole Baumgarth
- Center for Comparative MedicineUniversity of California, DavisDavisUnited States
| | - Gregory M Barton
- Department of Molecular and Cell BiologyUniversity of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
183
|
Weigert A, Olesch C, Brüne B. Sphingosine-1-Phosphate and Macrophage Biology-How the Sphinx Tames the Big Eater. Front Immunol 2019; 10:1706. [PMID: 31379883 PMCID: PMC6658986 DOI: 10.3389/fimmu.2019.01706] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022] Open
Abstract
The sphingolipid sphingosine-1-phosphate (S1P) is produced by sphingosine kinases to either signal through intracellular targets or to activate a family of specific G-protein-coupled receptors (S1PR). S1P levels are usually low in peripheral tissues compared to the vasculature, forming a gradient that mediates lymphocyte trafficking. However, S1P levels rise during inflammation in peripheral tissues, thereby affecting resident or recruited immune cells, including macrophages. As macrophages orchestrate initiation and resolution of inflammation, the sphingosine kinase/S1P/S1P-receptor axis emerges as an important determinant of macrophage function in the pathogenesis of inflammatory diseases such as cancer, atherosclerosis, and infection. In this review, we therefore summarize the current knowledge how S1P affects macrophage biology.
Collapse
Affiliation(s)
- Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Catherine Olesch
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany.,Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
184
|
Sheng YR, Hu WT, Wei CY, Tang LL, Liu YK, Liu YY, Qiu JP, Li DJ, Zhu XY. Insights of efferocytosis in normal and pathological pregnancy. Am J Reprod Immunol 2019; 82:e13088. [PMID: 30614132 DOI: 10.1111/aji.13088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/21/2018] [Accepted: 12/31/2018] [Indexed: 12/18/2022] Open
Abstract
Efferocytosis, which is known as the phagocytic clearance of dying cells by professional as well as non-professional phagocytes, including a great number of intracellular/extracellular factors and signals, is interrelated with the immune system, contributing to local and systemic homeostasis, especially in tissues with high constitutive rates of apoptosis. Accumulating studies have indicated that immune dysregulation is associated with the pathogenesis of the female reproductive system, which causes preeclampsia (PE), recurrent spontaneous abortion (RSA), ruptured ectopic pregnancy, and so on. And some studies have revealed the pleiotropic and essential role of efferocytosis in these obstetrical disorders. More specifically, the occurrence and development of these diseases were in connection with some efferocytosis-related factors and signals, such as C1q, MBL, and IL-33/ST2. In this review, we systematically review the diverse impacts of efferocytosis in immune system and discuss its relevance to normal and pathological pregnancy. These findings may instruct future basic researches as well as clinical applications of efferocytosis-related factors and signals as latent predictors or therapeutic targets on the obstetrical disorders.
Collapse
Affiliation(s)
- Yan-Ran Sheng
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Wen-Ting Hu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Chun-Yan Wei
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Ling-Li Tang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Yu-Kai Liu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Yu-Yin Liu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jian-Ping Qiu
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Xiao-Yong Zhu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
185
|
Abstract
Macrophages are critical mediators of tissue homeostasis, with tumours distorting this proclivity to stimulate proliferation, angiogenesis and metastasis. This had led to an interest in targeting macrophages in cancer, and preclinical studies have demonstrated efficacy across therapeutic modalities and tumour types. Much of the observed efficacy can be traced to the suppressive capacity of macrophages, driven by microenvironmental cues such as hypoxia and fibrosis. As a result, tumour macrophages display an ability to suppress T cell recruitment and function as well as to regulate other aspects of tumour immunity. With the increasing impact of cancer immunotherapy, macrophage targeting is now being evaluated in this context. Here, we discuss the results of clinical trials and the future of combinatorial immunotherapy.
Collapse
Affiliation(s)
- David G DeNardo
- Department of Medicine, ICCE Institute, Department of Pathology and Immunology, Siteman Cancer Center, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.
| | - Brian Ruffell
- Department of Immunology, Department of Breast Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
186
|
Abstract
Two recent Immunity papers provide new insight into efferocytosis by tissue-resident macrophages. Baratin et al. (2017) identify a resident macrophage population in the T cell zone of lymph nodes responsible for the silent uptake of vast numbers of apoptotic cells. Roberts et al. (2017) find that resident macrophages can be programmed by local tissue signals not to respond to the nucleic acid of apoptotic cells.
Collapse
Affiliation(s)
- Judith E Allen
- Faculty of Biology, Medicine, and Health, Division of Infection, Immunity, and Respiratory Medicine, University of Manchester, Manchester, M13 9PT UK.
| | - Dominik Rückerl
- Faculty of Biology, Medicine, and Health, Division of Infection, Immunity, and Respiratory Medicine, University of Manchester, Manchester, M13 9PT UK
| |
Collapse
|
187
|
Ma WT, Gao F, Gu K, Chen DK. The Role of Monocytes and Macrophages in Autoimmune Diseases: A Comprehensive Review. Front Immunol 2019; 10:1140. [PMID: 31178867 PMCID: PMC6543461 DOI: 10.3389/fimmu.2019.01140] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/07/2019] [Indexed: 12/19/2022] Open
Abstract
Monocytes (Mo) and macrophages (Mϕ) are key components of the innate immune system and are involved in regulation of the initiation, development, and resolution of many inflammatory disorders. In addition, these cells also play important immunoregulatory and tissue-repairing roles to decrease immune reactions and promote tissue regeneration. Several lines of evidence have suggested a causal link between the presence or activation of these cells and the development of autoimmune diseases. In addition, Mo or Mϕ infiltration in diseased tissues is a hallmark of several autoimmune diseases. However, the detailed contributions of these cells, whether they actually initiate disease or perpetuate disease progression, and whether their phenotype and functional alteration are merely epiphenomena are still unclear in many autoimmune diseases. Additionally, little is known about their heterogeneous populations in different autoimmune diseases. Elucidating the relevance of Mo and Mϕ in autoimmune diseases and the associated mechanisms could lead to the identification of more effective therapeutic strategies in the future.
Collapse
Affiliation(s)
- Wen-Tao Ma
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China.,School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Fei Gao
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kui Gu
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - De-Kun Chen
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
188
|
Svedberg FR, Brown SL, Krauss MZ, Campbell L, Sharpe C, Clausen M, Howell GJ, Clark H, Madsen J, Evans CM, Sutherland TE, Ivens AC, Thornton DJ, Grencis RK, Hussell T, Cunoosamy DM, Cook PC, MacDonald AS. The lung environment controls alveolar macrophage metabolism and responsiveness in type 2 inflammation. Nat Immunol 2019; 20:571-580. [PMID: 30936493 PMCID: PMC8381729 DOI: 10.1038/s41590-019-0352-y] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 02/14/2019] [Indexed: 02/06/2023]
Abstract
Fine control of macrophage activation is needed to prevent inflammatory disease, particularly at barrier sites such as the lungs. However, the dominant mechanisms that regulate the activation of pulmonary macrophages during inflammation are poorly understood. We found that alveolar macrophages (AlvMs) were much less able to respond to the canonical type 2 cytokine IL-4, which underpins allergic disease and parasitic worm infections, than macrophages from lung tissue or the peritoneal cavity. We found that the hyporesponsiveness of AlvMs to IL-4 depended upon the lung environment but was independent of the host microbiota or the lung extracellular matrix components surfactant protein D (SP-D) and mucin 5b (Muc5b). AlvMs showed severely dysregulated metabolism relative to that of cavity macrophages. After removal from the lungs, AlvMs regained responsiveness to IL-4 in a glycolysis-dependent manner. Thus, impaired glycolysis in the pulmonary niche regulates AlvM responsiveness during type 2 inflammation.
Collapse
Affiliation(s)
- Freya R Svedberg
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Laboratory of Myeloid Cell Ontogeny and Functional Specialisation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sheila L Brown
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Maria Z Krauss
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Laura Campbell
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Catherine Sharpe
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Maryam Clausen
- AstraZeneca, Discovery Sciences IMED, Gothenburg, Sweden
| | - Gareth J Howell
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Howard Clark
- Department of Child Health, Division of Clinical and Experimental Sciences, Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
- National Institute for Health Research, Southampton Respiratory Biomedical Research Unit, Southampton Centre for Biomedical Research, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jens Madsen
- Department of Child Health, Division of Clinical and Experimental Sciences, Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
- National Institute for Health Research, Southampton Respiratory Biomedical Research Unit, Southampton Centre for Biomedical Research, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Christopher M Evans
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Tara E Sutherland
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Alasdair C Ivens
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - David J Thornton
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Richard K Grencis
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Tracy Hussell
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Peter C Cook
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
189
|
Challa DK, Wang X, Montoyo HP, Velmurugan R, Ober RJ, Ward ES. Neonatal Fc receptor expression in macrophages is indispensable for IgG homeostasis. MAbs 2019; 11:848-860. [PMID: 30964743 PMCID: PMC6601554 DOI: 10.1080/19420862.2019.1602459] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The maintenance of the homeostasis of immunoglobulin G (IgG) represents a fundamental aspect of humoral immunity that has direct relevance to the successful delivery of antibody-based therapeutics. The ubiquitously expressed neonatal Fc receptor (FcRn) salvages IgG from cellular degradation following pinocytic uptake into cells, conferring prolonged in vivo persistence on IgG. However, the cellular sites of FcRn function are poorly defined. Pinocytic uptake is a prerequisite for FcRn-mediated IgG salvage, prompting us to investigate the consequences of IgG uptake and catabolism by macrophages, which represent both abundant and highly pinocytic cells in the body. Site-specific deletion of FcRn to generate mice harboring FcRn-deficient macrophages results in IgG hypercatabolism and ~threefold reductions in serum IgG levels, whereas these effects were not observed in mice that lack functional FcRn in B cells and dendritic cells. Consistent with the degradative activity of FcRn-deficient macrophages, depletion of these cells in FcRn-deficient mice leads to increased persistence and serum levels of IgG. These studies demonstrate a pivotal role for FcRn-mediated salvage in compensating for the high pinocytic and degradative activities of macrophages to maintain IgG homeostasis.
Collapse
Affiliation(s)
- Dilip K Challa
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA
| | - Xiaoli Wang
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA
| | - Héctor Pérez Montoyo
- b Department of Immunology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Ramraj Velmurugan
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA
| | - Raimund J Ober
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA.,c Department of Biomedical Engineering , Texas A&M University , College Station , TX , USA.,d Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine , University of Southampton , Southampton , UK
| | - E Sally Ward
- a Department of Molecular and Cellular Medicine , Texas A&M University Health Science Center , College Station , TX , USA.,d Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine , University of Southampton , Southampton , UK.,e Department of Microbial Pathogenesis and Immunology , Texas A&M University Health Science Center , Bryan , TX , USA
| |
Collapse
|
190
|
Galloway DA, Phillips AEM, Owen DRJ, Moore CS. Phagocytosis in the Brain: Homeostasis and Disease. Front Immunol 2019; 10:790. [PMID: 31040847 PMCID: PMC6477030 DOI: 10.3389/fimmu.2019.00790] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/26/2019] [Indexed: 12/28/2022] Open
Abstract
Microglia are resident macrophages of the central nervous system and significantly contribute to overall brain function by participating in phagocytosis during development, homeostasis, and diseased states. Phagocytosis is a highly complex process that is specialized for the uptake and removal of opsonized and non-opsonized targets, such as pathogens, apoptotic cells, and cellular debris. While the role of phagocytosis in mediating classical innate and adaptive immune responses has been known for decades, it is now appreciated that phagocytosis is also critical throughout early neural development, homeostasis, and initiating repair mechanisms. As such, modulating phagocytic processes has provided unexplored avenues with the intent of developing novel therapeutics that promote repair and regeneration in the CNS. Here, we review the functional consequences that phagocytosis plays in both the healthy and diseased CNS, and summarize how phagocytosis contributes to overall pathophysiological mechanisms involved in brain injury and repair.
Collapse
Affiliation(s)
- Dylan A Galloway
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Alexandra E M Phillips
- Division of Brain Sciences, Department of Medicine Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - David R J Owen
- Division of Brain Sciences, Department of Medicine Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
191
|
MacNabb BW, Kline DE, Albright AR, Chen X, Leventhal DS, Savage PA, Kline J. Negligible Role for Deletion Mediated by cDC1 in CD8 + T Cell Tolerance. THE JOURNAL OF IMMUNOLOGY 2019; 202:2628-2635. [PMID: 30902900 DOI: 10.4049/jimmunol.1801621] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/05/2019] [Indexed: 11/19/2022]
Abstract
Deletion of CD8+ T cells by dendritic cells (DCs) is recognized as a critical mechanism of immune tolerance to self-antigens. Although DC-mediated peripheral deletion of autoreactive CD8+ T cells has been demonstrated using T cells reactive to model Ags, its role in shaping the naturally occurring polyclonal CD8+ T cell repertoire has not been defined. Using Batf3-/- mice lacking cross-presenting CD8α+ and CD103+ DCs (also known as type 1 conventional [cDC1]), we demonstrate that peripheral deletion of CD8+ T cells reactive to a model tissue Ag is dependent on cDC1. However, endogenous CD8+ T cells from the periphery of Batf3-/- mice do not exhibit heightened self-reactivity, and deep TCR sequencing of CD8+ T cells from Batf3-/- and Batf3+/+ mice reveals that cDC1 have a minimal impact on shaping the peripheral CD8+ T cell repertoire. Thus, although evident in reductionist systems, deletion of polyclonal self-specific CD8+ T cells by cDC1 plays a negligible role in enforcing tolerance to natural self-ligands.
Collapse
Affiliation(s)
| | - Douglas E Kline
- Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - Annie R Albright
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Xiufen Chen
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Daniel S Leventhal
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637; and
| | - Peter A Savage
- Committee on Immunology, University of Chicago, Chicago, IL 60637.,Committee on Cancer Biology, University of Chicago, Chicago, IL 60637; and.,Department of Pathology, University of Chicago, Chicago, IL 60637
| | - Justin Kline
- Committee on Immunology, University of Chicago, Chicago, IL 60637; .,Department of Medicine, University of Chicago, Chicago, IL 60637.,Committee on Cancer Biology, University of Chicago, Chicago, IL 60637; and
| |
Collapse
|
192
|
Sima C, Viniegra A, Glogauer M. Macrophage immunomodulation in chronic osteolytic diseases-the case of periodontitis. J Leukoc Biol 2019; 105:473-487. [PMID: 30452781 PMCID: PMC6386606 DOI: 10.1002/jlb.1ru0818-310r] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022] Open
Abstract
Periodontitis (PD) is a chronic osteolytic disease that shares pathogenic inflammatory features with other conditions associated with nonresolving inflammation. A hallmark of PD is inflammation-mediated alveolar bone loss. Myeloid cells, in particular polymorphonuclear neutrophils (PMN) and macrophages (Mac), are essential players in PD by control of gingival biofilm pathogenicity, activation of adaptive immunity, as well as nonresolving inflammation and collateral tissue damage. Despite mounting evidence of significant innate immune implications to PD progression and healing after therapy, myeloid cell markers and targets for immune modulation have not been validated for clinical use. The remarkable plasticity of monocytes/Mac in response to local activation factors enables these cells to play central roles in inflammation and restoration of tissue homeostasis and provides opportunities for biomarker and therapeutic target discovery for management of chronic inflammatory conditions, including osteolytic diseases such as PD and arthritis. Along a wide spectrum of activation states ranging from proinflammatory to pro-resolving, Macs respond to environmental changes in a site-specific manner in virtually all tissues. This review summarizes the existing evidence on Mac immunomodulation therapies for osteolytic diseases in the broader context of conditions associated with nonresolving inflammation, and discusses osteoimmune implications of Macs in PD.
Collapse
Affiliation(s)
- Corneliu Sima
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Ana Viniegra
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Michael Glogauer
- Dental Research Institute, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
193
|
Paludan SR, Reinert LS, Hornung V. DNA-stimulated cell death: implications for host defence, inflammatory diseases and cancer. Nat Rev Immunol 2019; 19:141-153. [PMID: 30644449 PMCID: PMC7311199 DOI: 10.1038/s41577-018-0117-0] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The immune system detects disturbances in homeostasis that occur during infection, sterile tissue damage and cancer. This initiates immune responses that seek to eliminate the trigger of immune activation and to re-establish homeostasis. At the same time, these mechanisms can also play a crucial role in the progression of disease. The occurrence of DNA in the cytosol constitutes a potent trigger for the innate immune system, governing the production of key inflammatory cytokines such as type I interferons and IL-1β. More recently, it has become clear that cytosolic DNA also triggers other biological responses, including various forms of programmed cell death. In this article, we review the emerging literature on the pathways governing DNA-stimulated cell death and the current knowledge on how these processes shape immune responses to exogenous and endogenous challenges.
Collapse
Affiliation(s)
- Søren R Paludan
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark.
- Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Line S Reinert
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Veit Hornung
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany.
- Center for Integrated Protein Science (CIPSM), Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
194
|
Horuluoglu B, Bayik D, Kayraklioglu N, Goguet E, Kaplan MJ, Klinman DM. PAM3 supports the generation of M2-like macrophages from lupus patient monocytes and improves disease outcome in murine lupus. J Autoimmun 2019; 99:24-32. [PMID: 30679006 DOI: 10.1016/j.jaut.2019.01.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/01/2023]
Abstract
Systematic Lupus Erythematosus (SLE) is an autoimmune syndrome of unclear etiology. While T and B cell abnormalities contribute to disease pathogenesis, recent work suggests that inflammatory M1-like macrophages also play a role. Previous work showed that the TLR2/1 agonist PAM3CSK4 (PAM3) could stimulate normal human monocytes to preferentially differentiate into immunosuppressive M2-like rather than inflammatory M1-like macrophages. This raised the possibility of PAM3 being used to normalize the M1:M2 ratio in SLE. Consistent with that possibility, monocytes from lupus patients differentiated into M2-like macrophages when treated with PAM3 in vitro. Furthermore, lupus-prone NZB x NZW F1 mice responded similarly to weekly PAM3 treatment. Normalization of the M2 macrophage frequency was associated with delayed disease progression, decreased autoantibody and inflammatory cytokine synthesis, reduced proteinuria and prolonged survival in NZB x NZW F1 mice. The ability of PAM3 to bias monocyte differentiation in favor of immunosuppressive macrophages may represent a novel approach to the therapy of SLE.
Collapse
Affiliation(s)
- Begum Horuluoglu
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA; Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Defne Bayik
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA
| | - Neslihan Kayraklioglu
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA
| | - Emilie Goguet
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Dennis M Klinman
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA.
| |
Collapse
|
195
|
|
196
|
Nobs SP, Kayhan M, Kopf M. GM-CSF intrinsically controls eosinophil accumulation in the setting of allergic airway inflammation. J Allergy Clin Immunol 2018; 143:1513-1524.e2. [PMID: 30244025 DOI: 10.1016/j.jaci.2018.08.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/09/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Eosinophils are a therapeutic target in asthmatic patients, and GM-CSF has been suggested to control various aspects of eosinophil biology, including development, function, and survival. However, to date, the role of GM-CSF signaling in eosinophils in vivo is largely unclear. OBJECTIVE We sought to elucidate the role of GM-CSF signaling in asthmatic inflammation. METHODS Wild-type and GM-CSF receptor α (Csf2ra)-deficient mice reconstituted with Csf2ra-proficient alveolar macrophages were subjected to different models of airway inflammation to evaluate the effect of GM-CSF signaling deficiency on asthmatic inflammation in general and on eosinophils in particular. RESULTS We demonstrate that GM-CSF signaling, although being largely dispensable for eosinophil development at steady state, intrinsically promotes accumulation of eosinophils in the lung during allergic airway inflammation. In contrast, chitin-induced eosinophil accumulation in the peritoneal cavity occurs independent of GM-CSF, indicating organ specificity. We show that GM-CSF induces chemokinesis and promotes eosinophil survival in vitro, which likely contribute to eosinophil accumulation in the airways in vivo. CONCLUSION GM-CSF intrinsically promotes eosinophil accumulation in the setting of pulmonary allergic inflammation.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland; the Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merve Kayhan
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
197
|
Price AE, Shamardani K, Lugo KA, Deguine J, Roberts AW, Lee BL, Barton GM. A Map of Toll-like Receptor Expression in the Intestinal Epithelium Reveals Distinct Spatial, Cell Type-Specific, and Temporal Patterns. Immunity 2018; 49:560-575.e6. [PMID: 30170812 PMCID: PMC6152941 DOI: 10.1016/j.immuni.2018.07.016] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/21/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022]
Abstract
Signaling by Toll-like receptors (TLRs) on intestinal epithelial cells (IECs) is critical for intestinal homeostasis. To visualize epithelial expression of individual TLRs in vivo, we generated five strains of reporter mice. These mice revealed that TLR expression varied dramatically along the length of the intestine. Indeed, small intestine (SI) IECs expressed low levels of multiple TLRs that were highly expressed by colonic IECs. TLR5 expression was restricted to Paneth cells in the SI epithelium. Intestinal organoid experiments revealed that TLR signaling in Paneth cells or colonic IECs induced a core set of host defense genes, but this set did not include antimicrobial peptides, which instead were induced indirectly by inflammatory cytokines. This comprehensive blueprint of TLR expression and function in IECs reveals unexpected diversity in the responsiveness of IECs to microbial stimuli, and together with the associated reporter strains, provides a resource for further study of innate immunity.
Collapse
Affiliation(s)
- April E Price
- Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Kiarash Shamardani
- Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Kyler A Lugo
- Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Jacques Deguine
- Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Allison W Roberts
- Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Bettina L Lee
- Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Gregory M Barton
- Division of Immunology & Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
198
|
Asano K, Kikuchi K, Tanaka M. CD169 macrophages regulate immune responses toward particulate materials in the circulating fluid. J Biochem 2018; 164:77-85. [PMID: 29905851 DOI: 10.1093/jb/mvy050] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022] Open
Abstract
Tissue macrophages comprise heterogeneous subsets that differ in localization, phenotype and ontogeny. They acquire tissue-specific phenotype in order to maintain normal tissue physiology. This review summarizes the current knowledge about the functions of CD169-positive macrophage subset residing in the lymphoid organs and intestinal tract. Strategically positioned at the interface between tissue and circulating fluid, CD169+ macrophages in the lymphoid organs capture blood- and lymph-borne particulate materials. Antigen information relayed by CD169+ macrophages to neighbouring immune cells is important for enhancement of antimicrobial and antitumour immunity as well as induction of tolerance. In the intestinal tract, CD169+ macrophages localize distantly from epithelial border. Following mucosal injury, they exacerbate inflammation by producing CCL8 that recruits inflammatory monocytes. As such, a better understanding of CD169+ macrophage phenotypes may enable the design of tissue-specific therapies for both immunological and non-immunological diseases.
Collapse
Affiliation(s)
- Kenichi Asano
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Kenta Kikuchi
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Masato Tanaka
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
199
|
The biology of serous cavity macrophages. Cell Immunol 2018; 330:126-135. [DOI: 10.1016/j.cellimm.2018.01.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/03/2018] [Indexed: 12/19/2022]
|
200
|
Alivernini S, Tolusso B, Ferraccioli G, Gremese E, Kurowska-Stolarska M, McInnes IB. Driving chronicity in rheumatoid arthritis: perpetuating role of myeloid cells. Clin Exp Immunol 2018; 193:13-23. [PMID: 29315512 PMCID: PMC6038003 DOI: 10.1111/cei.13098] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 12/11/2022] Open
Abstract
Acute inflammation is a complex and tightly regulated homeostatic process that includes leucocyte migration from the vasculature into tissues to eliminate the pathogen/injury, followed by a pro‐resolving response promoting tissue repair. However, if inflammation is uncontrolled as in chronic diseases such as rheumatoid arthritis (RA), it leads to tissue damage and disability. Synovial tissue inflammation in RA patients is maintained by sustained activation of multiple inflammatory positive‐feedback regulatory pathways in a variety of cells, including myeloid cells. In this review, we will highlight recent evidence uncovering biological mechanisms contributing to the aberrant activation of myeloid cells that contributes to perpetuation of inflammation in RA, and discuss emerging data on anti‐inflammatory mediators contributing to sustained remission that may inform a novel category of therapeutic targets.
Collapse
Affiliation(s)
- S Alivernini
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - B Tolusso
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - G Ferraccioli
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - E Gremese
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - M Kurowska-Stolarska
- Institute of Infection, Immunity and Inflammation, University of Glasgow.,Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, UK
| | - I B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow.,Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, UK
| |
Collapse
|