1
|
Hao G, Han S, Xiao Z, Shen J, Zhao Y, Hao Q. Synovial mast cells and osteoarthritis: Current understandings and future perspectives. Heliyon 2024; 10:e41003. [PMID: 39720069 PMCID: PMC11665477 DOI: 10.1016/j.heliyon.2024.e41003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 10/29/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent joint disease worldwide that significantly impacts the quality of life of individuals, particularly those in middle-aged and elderly populations. OA was initially considered as non-inflammatory arthritis, but recent studies have identified a substantial number of immune responses in OA, leading to the recognition of inflammation as a key factor in its pathogenesis. An increasing number of studies have found that mast cell (MC) and MC-secreted inflammatory mediators and cytokines are notably increased in the synovial fluid of OA patients, indicating a potential association between MCs and the onset and progression of synovial inflammation. The present review aims to summarize the significance and mechanism of MCs in the pathogenesis of OA. Meanwhile, we also discuss the clinical potential of using MCs as therapeutic target for OA therapy. Modulating the activities of MCs or the mediators of MCs in the synovial fluid inflammatory microenvironment will be promising new options for the treatment of OA.
Collapse
Affiliation(s)
- Guanghui Hao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shanqian Han
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Southwest Medical University, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Qi Hao
- Department of Joint Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- The Third People's Hospital of Longmatan District, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Farinelli L, D'Angelo F, Ciccullo C, Manzotti S, Gigante A. A significant difference of synovial mast cells in synovium from rotator cuff arthropathy compared to rotator cuff tears: A histological pilot study. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100503. [PMID: 39156865 PMCID: PMC11326889 DOI: 10.1016/j.ocarto.2024.100503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Abstract
Objective Aim of the present study was to compare the presence of Mast Cells (MCs) in synovial samples from gleno-humeral osteoarthritis (OA) and from control group. Methods Synovial tissue samples were obtained during arthroplasty from 23 patients with gleno-humeral OA due to rotator cuff arthropathy (RCA) and from 20 patients without OA, constituting OA group and control group respectively. Before surgery self-reported pain was assessed using VAS score and OSS was used to value functional ability. Shoulder radiograph (Antero-posterior, Y-view and Grashey views) was evaluated by musculoskeletal radiologist and graded according to modified Samilson-Prieto classification.Synovial tissue, obtained during arthroplasty and arthroscopic procedure, was prepared to immunohistochemical analysis with anti-CD31 and anti-CD117 antibodies, to detect respectively endothelial cells and MCs at 40x magnification. Synovitis scores have been assessed. Under the control of the image processing system the distribution and the total number of vessels and MCs were determined. Results The numbers of MCs and the area fraction (20x magnification) occupied by them were significantly higher in OA samples than in control tissue. The synovitis score was higher in OA patients with a positive correlation. Vessels number and area fraction were higher in OA patients than in controls. Analysis of MC number in relation to clinical data indicated positive correlation with the VAS score. Conclusions The distribution of MCs on synovium significantly differ between OA and control groups. Despite the design of the study could not conclude the cause-effect relationship, the presence of MCs might have role in OA pathogenesis. Level of evidence Histological study.
Collapse
Affiliation(s)
- Luca Farinelli
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco D'Angelo
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Carlo Ciccullo
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Sandra Manzotti
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Gigante
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
- IRCCS INRCA, Ancona, Italy
| |
Collapse
|
3
|
He B, Liao Y, Tian M, Tang C, Tang Q, Ma F, Zhou W, Leng Y, Zhong D. Identification and verification of a novel signature that combines cuproptosis-related genes with ferroptosis-related genes in osteoarthritis using bioinformatics analysis and experimental validation. Arthritis Res Ther 2024; 26:100. [PMID: 38741149 DOI: 10.1186/s13075-024-03328-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Exploring the pathogenesis of osteoarthritis (OA) is important for its prevention, diagnosis, and treatment. Therefore, we aimed to construct novel signature genes (c-FRGs) combining cuproptosis-related genes (CRGs) with ferroptosis-related genes (FRGs) to explore the pathogenesis of OA and aid in its treatment. MATERIALS AND METHODS Differentially expressed c-FRGs (c-FDEGs) were obtained using R software. Enrichment analysis was performed and a protein-protein interaction (PPI) network was constructed based on these c-FDEGs. Then, seven hub genes were screened. Three machine learning methods and verification experiments were used to identify four signature biomarkers from c-FDEGs, after which gene set enrichment analysis, gene set variation analysis, single-sample gene set enrichment analysis, immune function analysis, drug prediction, and ceRNA network analysis were performed based on these signature biomarkers. Subsequently, a disease model of OA was constructed using these biomarkers and validated on the GSE82107 dataset. Finally, we analyzed the distribution of the expression of these c-FDEGs in various cell populations. RESULTS A total of 63 FRGs were found to be closely associated with 11 CRGs, and 40 c-FDEGs were identified. Bioenrichment analysis showed that they were mainly associated with inflammation, external cellular stimulation, and autophagy. CDKN1A, FZD7, GABARAPL2, and SLC39A14 were identified as OA signature biomarkers, and their corresponding miRNAs and lncRNAs were predicted. Finally, scRNA-seq data analysis showed that the differentially expressed c-FRGs had significantly different expression distributions across the cell populations. CONCLUSION Four genes, namely CDKN1A, FZD7, GABARAPL2, and SLC39A14, are excellent biomarkers and prospective therapeutic targets for OA.
Collapse
Affiliation(s)
- Baoqiang He
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, No. 25 Taping Street, Lu Zhou City, China
- Southwest Medical University, Lu Zhou City, China
| | - Yehui Liao
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, No. 25 Taping Street, Lu Zhou City, China
| | - Minghao Tian
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, No. 25 Taping Street, Lu Zhou City, China
| | - Chao Tang
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, No. 25 Taping Street, Lu Zhou City, China
| | - Qiang Tang
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, No. 25 Taping Street, Lu Zhou City, China
| | - Fei Ma
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, No. 25 Taping Street, Lu Zhou City, China
| | - Wenyang Zhou
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, No. 25 Taping Street, Lu Zhou City, China
| | - Yebo Leng
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, No. 25 Taping Street, Lu Zhou City, China.
- Meishan Tianfu New Area People's Hospital, Meishan City, China.
| | - Dejun Zhong
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, No. 25 Taping Street, Lu Zhou City, China.
- Southwest Medical University, Lu Zhou City, China.
| |
Collapse
|
4
|
Gutowski Ł, Kanikowski S, Formanowicz D. Mast Cell Involvement in the Pathogenesis of Selected Musculoskeletal Diseases. Life (Basel) 2023; 13:1690. [PMID: 37629547 PMCID: PMC10455104 DOI: 10.3390/life13081690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
In recent years, there has been a noteworthy revival of interest in the function of mast cells (MCs) in the human body. It is now acknowledged that MCs impact a wide array of processes beyond just allergies, leading to a shift in research direction. Unfortunately, some earlier conclusions were drawn from animal models with flawed designs, particularly centered around the receptor tyrosine kinase (Kit) pathway. Consequently, several subsequent findings may have been unreliable. Thus, what is now required is a re-examination of these earlier findings. Nevertheless, the remaining data are fascinating and hold promise for a better comprehension of numerous diseases and the development of more effective therapies. As the field continues to progress, many intriguing issues warrant further investigation and analysis. For instance, exploring the bidirectional action of MCs in rheumatoid arthritis, understanding the extent of MCs' impact on symptoms associated with Ehlers-Danlos syndrome, and unraveling the exact role of the myofibroblast-mast cell-neuropeptides axis in the joint capsule during post-traumatic contractures are all captivating areas for exploration. Hence, in this review, we summarize current knowledge regarding the influence of MCs on the pathogenesis of selected musculoskeletal diseases, including rheumatoid arthritis, spondyloarthritis, psoriatic arthritis, gout, muscle and joint injuries, tendinopathy, heterotopic ossification, and Ehlers-Danlos syndrome. We believe that this review will provide in-depth information that can guide and inspire further research in this area.
Collapse
Affiliation(s)
- Łukasz Gutowski
- Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland;
| | - Szymon Kanikowski
- Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland;
| | - Dorota Formanowicz
- Department of Medical Chemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland;
- Department of Stem Cells and Regenerative Medicine, Institute of Natural Fibres and Medicinal Plants—National Research Institute, Kolejowa 2, 62-064 Plewiska, Poland
| |
Collapse
|
5
|
Loucks A, Maerz T, Hankenson K, Moeser A, Colbath A. The multifaceted role of mast cells in joint inflammation and arthritis. Osteoarthritis Cartilage 2023; 31:567-575. [PMID: 36682447 DOI: 10.1016/j.joca.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To review current knowledge surrounding the role of mast cells in joint inflammation and arthritis. METHOD Narrative review. RESULTS Mast cells (MCs) are commonly observed in the synovium of the joint, particularly surrounding blood vessels and nerve endings. Some studies have reported increased MC number and degranulation in patients with osteoarthritis (OA). In two studies, MCs were the only immune cell type found in higher concentrations in synovium of OA patients compared to rheumatoid arthritis patients. Activation of MCs in OA includes signaling pathways such as immunoglobulin E/Fc epsilon Receptor 1 (IgE/FcεR1), immunoglobulin G/Fc gamma receptor (IgG/FcγR), complement, and toll-like cell surface receptor-mediated signaling, resulting in context-dependent release of either pro-inflammatory and/or anti-inflammatory mediators within the joint. Activation of MCs results in the release of pro-inflammatory mediators that ultimately contribute to inflammation of the synovium, bone remodeling, and cartilage damage. However, some studies have proposed that MCs can also exhibit anti-inflammatory effects by secreting mediators that inactivate pro-inflammatory cytokines such as interleukin 6 (IL-6). CONCLUSIONS MCs may play a role in mediating synovial inflammation and OA progression. However, the mechanisms governing MC activation, the downstream pro- and/or anti-inflammatory effects, and their impact on osteoarthritis pathogenesis remains to be elucidated and requires extensive further study. Furthermore, it is important to establish the pathways of MC activation in OA to determine whether MCs exhibit varying phenotypes as a function of disease stage. Ultimately, such research is needed before understanding whether MCs could be targeted in OA treatments.
Collapse
Affiliation(s)
- A Loucks
- Department of Clinical Sciences, Cornell University, Ithaca, NY, USA.
| | - T Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - K Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - A Moeser
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA.
| | - A Colbath
- Department of Clinical Sciences, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
6
|
Loucks A, Maerz T, Hankenson K, Moeser A, Colbath A. WITHDRAWN: The Multifaceted Role of Mast Cells in Joint Inflammation and Arthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2022. [DOI: 10.1016/j.ocarto.2022.100309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
7
|
Farinelli L, Aquili A, Mattioli-Belmonte M, Manzotti S, D'Angelo F, Ciccullo C, Gigante A. Synovial mast cells from knee and hip osteoarthritis: histological study and clinical correlations. J Exp Orthop 2022; 9:13. [PMID: 35079910 PMCID: PMC8789998 DOI: 10.1186/s40634-022-00446-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/07/2022] [Indexed: 02/01/2023] Open
Abstract
PURPOSE The aim of this study was to investigate the presence of synovial mast cells (MCs) in hip and knee tissue from osteoarthritis (OA) patients and to correlate them with clinical and radiological data. METHODS Synovial tissue was obtained during arthroplasty from 60 patients, 30 with knee OA and 30 with hip OA. Control synovial tissue was obtained from 30 patients without OA, 15 undergoing above-knee amputation and 15 receiving a hip replacement for fracture. Before surgery, the radiographic findings were graded according to the Kellgren-Lawrence system and clinical data including pain (VAS) and functional information (KOOS and HOOS) was collected. The tissue was stained with hematoxylin-eosin and toluidine blue for histochemistry and incubated with CD117 and CD31 antibodies for immunohistochemistry. MC and vessel number and synovitis score were determined in all samples. RESULTS Mean MC number, synovitis score and vessel number were significantly higher in the OA samples (p < 0.05) than in control tissue. MC number correlated with the synovitis score and disease severity in both patient groups. CONCLUSIONS The prevalence of MCs in synovium from OA patients and their association with synovial inflammation and pain suggest a role for them in OA pathophysiology.
Collapse
Affiliation(s)
- L Farinelli
- Clinical Orthopaedics, Department of Clinical and Molecular Science DISCLIMO, Università Politecnica Delle Marche, Ancona, Italy
| | - A Aquili
- Clinical Orthopaedics, Department of Clinical and Molecular Science DISCLIMO, Università Politecnica Delle Marche, Ancona, Italy
| | - M Mattioli-Belmonte
- Department of Clinical and Molecular Science DISCLIMO, Università Politecnica Delle Marche, Ancona, Italy
| | - S Manzotti
- Clinical Orthopaedics, Department of Clinical and Molecular Science DISCLIMO, Università Politecnica Delle Marche, Ancona, Italy
| | - F D'Angelo
- Clinical Orthopaedics, Department of Clinical and Molecular Science DISCLIMO, Università Politecnica Delle Marche, Ancona, Italy
| | - C Ciccullo
- Clinical Orthopaedics, Department of Clinical and Molecular Science DISCLIMO, Università Politecnica Delle Marche, Ancona, Italy
| | - A Gigante
- Clinical Orthopaedics, Department of Clinical and Molecular Science DISCLIMO, Università Politecnica Delle Marche, Ancona, Italy.
| |
Collapse
|
8
|
Chalayer E, Gramont B, Zekre F, Goguyer-Deschaumes R, Waeckel L, Grange L, Paul S, Chung AW, Killian M. Fc receptors gone wrong: A comprehensive review of their roles in autoimmune and inflammatory diseases. Autoimmun Rev 2021; 21:103016. [PMID: 34915182 DOI: 10.1016/j.autrev.2021.103016] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/08/2021] [Indexed: 12/16/2022]
Abstract
Systemic autoimmune and inflammatory diseases have a complex and only partially known pathophysiology with various abnormalities involving all the components of the immune system. Among these components, antibodies, and especially autoantibodies are key elements contributing to autoimmunity. The interaction of antibody fragment crystallisable (Fc) and several distinct receptors, namely Fc receptors (FcRs), have gained much attention during the recent years, with possible major therapeutic perspectives for the future. The aim of this review is to comprehensively describe the known roles for FcRs (activating and inhibitory FcγRs, neonatal FcR [FcRn], FcαRI, FcεRs, Ro52/tripartite motif containing 21 [Ro52/TRIM21], FcδR, and the novel Fc receptor-like [FcRL] family) in systemic autoimmune and inflammatory disorders, namely rheumatoid arthritis, Sjögren's syndrome, systemic lupus erythematosus, systemic sclerosis, idiopathic inflammatory myopathies, mixed connective tissue disease, Crohn's disease, ulcerative colitis, immunoglobulin (Ig) A vasculitis, Behçet's disease, Kawasaki disease, IgG4-related disease, immune thrombocytopenia, autoimmune hemolytic anemia, antiphospholipid syndrome and heparin-induced thrombocytopenia.
Collapse
Affiliation(s)
- Emilie Chalayer
- Department of Hematology and Cell Therapy, Institut de Cancérologie Lucien Neuwirth, Saint-Etienne, France; INSERM U1059-Sainbiose, dysfonction vasculaire et hémostase, Université de Lyon, Saint-Etienne, France
| | - Baptiste Gramont
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Franck Zekre
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Pediatrics, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Roman Goguyer-Deschaumes
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France
| | - Louis Waeckel
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Immunology, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Lucile Grange
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Immunology, Saint-Etienne University Hospital, Saint-Etienne, France
| | - Amy W Chung
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Martin Killian
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université de Lyon, Université Jean Monnet, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR530, F42023 Saint-Etienne, France; Department of Internal Medicine, Saint-Etienne University Hospital, Saint-Etienne, France.
| |
Collapse
|
9
|
Noto CN, Hoft SG, DiPaolo RJ. Mast Cells as Important Regulators in Autoimmunity and Cancer Development. Front Cell Dev Biol 2021; 9:752350. [PMID: 34712668 PMCID: PMC8546116 DOI: 10.3389/fcell.2021.752350] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/17/2021] [Indexed: 01/04/2023] Open
Abstract
Mast cells are an essential part of the immune system and are best known as important modulators of allergic and anaphylactic immune responses. Upon activation, mast cells release a multitude of inflammatory mediators with various effector functions that can be both protective and damage-inducing. Mast cells can have an anti-inflammatory or pro-inflammatory immunological effect and play important roles in regulating autoimmune diseases including rheumatoid arthritis, type 1 diabetes, and multiple sclerosis. Importantly, chronic inflammation and autoimmunity are linked to the development of specific cancers including pancreatic cancer, prostate cancer, colorectal cancer, and gastric cancer. Inflammatory mediators released from activated mast cells regulate immune responses and promote vascular permeability and the recruitment of immune cells to the site of inflammation. Mast cells are present in increased numbers in tissues affected by autoimmune diseases as well as in tumor microenvironments where they co-localize with T regulatory cells and T effector cells. Mast cells can regulate immune responses by expressing immune checkpoint molecules on their surface, releasing anti-inflammatory cytokines, and promoting vascularization of solid tumor sites. As a result of these immune modulating activities, mast cells have disease-modifying roles in specific autoimmune diseases and cancers. Therefore, determining how to regulate the activities of mast cells in different inflammatory and tumor microenvironments may be critical to discovering potential therapeutic targets to treat autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Christine N Noto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Stella G Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Richard J DiPaolo
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
10
|
Ogasawara H, Noguchi M. Therapeutic Potential of MRGPRX2 Inhibitors on Mast Cells. Cells 2021; 10:cells10112906. [PMID: 34831128 PMCID: PMC8616451 DOI: 10.3390/cells10112906] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022] Open
Abstract
Mast cells (MCs) act as primary effectors in inflammatory and allergic reactions by releasing intracellularly-stored inflammatory mediators in diseases. The two major pathways for MC activation are known to be immunoglobulin E (IgE)-dependent and -independent. Although IgE-dependent signaling is the main pathway to MC activation, IgE-independent pathways have also been found to serve pivotal roles in the pathophysiology of various inflammatory conditions. Recent studies have shown that human and mouse MCs express several regulatory receptors such as toll-like receptors (TLRs), CD48, C300a, and GPCRs, including mas-related GPCR-X2 (MRGPRX2). MRGPRX2 has been reported as a novel GPCR that is expressed in MCs activated by basic secretagogues, neurokinin peptides, host defense antimicrobial peptides, and small molecule compounds (e.g., neuromuscular blocking agents) and leads to MC degranulation and eicosanoids release under in vitro experimental condition. Functional analyses of MRGPRX2 and Mrgprb2 (mouse ortholog) indicate that MRGPRX2 is involved in MC hypersensitivity reactions causing neuroinflammation such as postoperative pain, type 2 inflammation, non-histaminergic itch, and drug-induced anaphylactic-like reactions. In this review, we discuss the roles in innate immunity through functional studies on MRGPRX2-mediated IgE-independent MC activation and also the therapeutic potential of MRGPRX2 inhibitors on allergic and inflammatory diseases.
Collapse
Affiliation(s)
- Hiroyuki Ogasawara
- Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Yokohama 236-0004, Japan;
- Correspondence: ; Tel.: +81-45-786-7690
| | - Masato Noguchi
- Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Yokohama 236-0004, Japan;
- Office of Research Development and Sponsored Projects, Shinanomachi Campus, Keio University, Tokyo 160-8582, Japan
| |
Collapse
|
11
|
Saxena Y, Routh S, Mukhopadhaya A. Immunoporosis: Role of Innate Immune Cells in Osteoporosis. Front Immunol 2021; 12:687037. [PMID: 34421899 PMCID: PMC8374941 DOI: 10.3389/fimmu.2021.687037] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis or porous bone disorder is the result of an imbalance in an otherwise highly balanced physiological process known as 'bone remodeling'. The immune system is intricately involved in bone physiology as well as pathologies. Inflammatory diseases are often correlated with osteoporosis. Inflammatory mediators such as reactive oxygen species (ROS), and pro-inflammatory cytokines and chemokines directly or indirectly act on the bone cells and play a role in the pathogenesis of osteoporosis. Recently, Srivastava et al. (Srivastava RK, Dar HY, Mishra PK. Immunoporosis: Immunology of Osteoporosis-Role of T Cells. Frontiers in immunology. 2018;9:657) have coined the term "immunoporosis" to emphasize the role of immune cells in the pathology of osteoporosis. Accumulated pieces of evidence suggest both innate and adaptive immune cells contribute to osteoporosis. However, innate cells are the major effectors of inflammation. They sense various triggers to inflammation such as pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), cellular stress, etc., thus producing pro-inflammatory mediators that play a critical role in the pathogenesis of osteoporosis. In this review, we have discussed the role of the innate immune cells in great detail and divided these cells into different sections in a systemic manner. In the beginning, we talked about cells of the myeloid lineage, including macrophages, monocytes, and dendritic cells. This group of cells explicitly influences the skeletal system by the action of production of pro-inflammatory cytokines and can transdifferentiate into osteoclast. Other cells of the myeloid lineage, such as neutrophils, eosinophils, and mast cells, largely impact osteoporosis via the production of pro-inflammatory cytokines. Further, we talked about the cells of the lymphoid lineage, including natural killer cells and innate lymphoid cells, which share innate-like properties and play a role in osteoporosis. In addition to various innate immune cells, we also discussed the impact of classical pro-inflammatory cytokines on osteoporosis. We also highlighted the studies regarding the impact of physiological and metabolic changes in the body, which results in chronic inflammatory conditions such as ageing, ultimately triggering osteoporosis.
Collapse
Affiliation(s)
- Yogesh Saxena
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Sanjeev Routh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Arunika Mukhopadhaya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| |
Collapse
|
12
|
Li Z, Huang Z, Bai L. Cell Interplay in Osteoarthritis. Front Cell Dev Biol 2021; 9:720477. [PMID: 34414194 PMCID: PMC8369508 DOI: 10.3389/fcell.2021.720477] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a common chronic disease and a significant health concern that needs to be urgently solved. OA affects the cartilage and entire joint tissues, including the subchondral bone, synovium, and infrapatellar fat pads. The physiological and pathological changes in these tissues affect the occurrence and development of OA. Understanding complex crosstalk among different joint tissues and their roles in OA initiation and progression is critical in elucidating the pathogenic mechanism of OA. In this review, we begin with an overview of the role of chondrocytes, synovial cells (synovial fibroblasts and macrophages), mast cells, osteoblasts, osteoclasts, various stem cells, and engineered cells (induced pluripotent stem cells) in OA pathogenesis. Then, we discuss the various mechanisms by which these cells communicate, including paracrine signaling, local microenvironment, co-culture, extracellular vesicles (exosomes), and cell tissue engineering. We particularly focus on the therapeutic potential and clinical applications of stem cell-derived extracellular vesicles, which serve as modulators of cell-to-cell communication, in the field of regenerative medicine, such as cartilage repair. Finally, the challenges and limitations related to exosome-based treatment for OA are discussed. This article provides a comprehensive summary of key cells that might be targets of future therapies for OA.
Collapse
Affiliation(s)
- Zihao Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyu Huang
- Foreign Languages College, Shanghai Normal University, Shanghai, China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Bone marrow derived mast cells injected into the osteoarthritic knee joints of mice induced by sodium monoiodoacetate enhanced spontaneous pain through activation of PAR2 and action of extracellular ATP. PLoS One 2021; 16:e0252590. [PMID: 34086763 PMCID: PMC8177436 DOI: 10.1371/journal.pone.0252590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
Conditions that resemble osteoarthritis (OA) were produced by injection of sodium monoiodoacetate (MIA) into the knee joints of mice. Bone marrow derived mast cells (BMMCs) injected into the OA knee joints enhanced spontaneous pain. Since no spontaneous pain was observed when BMMCs were injected into the knee joints of control mice that had not been treated with MIA, BMMCs should be activated within the OA knee joints and release some pain-inducible factors. Protease activated receptor-2 (PAR2) antagonist (FSLLRY-NH2) almost abolished the pain-enhancing effects of BMMCs injected into the OA knee joints, suggesting that tryptase, a mast cell protease that is capable of activating PAR2, should be released from the injected BMMCs and enhance pain through activation of PAR2. When PAR2 agonist (SLIGKV-NH2) instead of BMMCs was injected into the OA knee joints, it was also enhanced pain. Apyrase, an ATP degrading enzyme, injected into the OA knee joints before BMMCs suppressed the pain enhanced by BMMCs. We showed that purinoceptors (P2X4 and P2X7) were expressed in BMMCs and that extracellular ATP stimulated the release of tryptase from BMMCs. These observations suggest that ATP may stimulate degranulation of BMMCs and thereby enhanced pain. BMMCs injected into the OA knee joints stimulated expression of IL-1β, IL-6, TNF-α, CCL2, and MMP9 genes in the infrapatellar fat pads, and PAR2 antagonist suppressed the stimulatory effects of BMMCs. Our study suggests that intermittent pain frequently observed in OA knee joints may be due, at least partly, to mast cells through activation of PAR2 and action of ATP, and that intraarticular injection of BMMCs into the OA knee joints may provide a useful experimental system for investigating molecular mechanisms by which pain is induced in OA knee joints.
Collapse
|
14
|
Kim KW, Kim BM, Won JY, Min HK, Lee KA, Lee SH, Kim HR. Regulation of osteoclastogenesis by mast cell in rheumatoid arthritis. Arthritis Res Ther 2021; 23:124. [PMID: 33882986 PMCID: PMC8059019 DOI: 10.1186/s13075-021-02491-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 03/23/2021] [Indexed: 12/30/2022] Open
Abstract
Background In the pathogenesis of rheumatoid arthritis (RA), the role of mast cells has not been revealed clearly. We aimed to define the inflammatory and tissue-destructive roles of mast cells in rheumatoid arthritis (RA). Methods Serum and synovial fluid (SF) concentration levels of tryptase, chymase, and histamine were quantified using ELISA. After activating mast cells using IL-33, the production of TNF-α, IL-1β, IL-6, IL-17, RANKL, and MMPs was determined using real-time PCR and ELISA. Osteoclastogenesis was assessed in CD14+ monocytes from peripheral blood and SF, which were cultured with IL-33-activated mast cells, by counting TRAP-positive multinucleated cells. Results The concentration levels of serum tryptase, chymase, and histamine and SF histamine were higher in patients with RA than in controls. FcεR1 and c-kit-positive mast cells were higher in RA synovium than in osteoarthritic (OA) synovium. Stimulation of mast cells by IL-33 increased the number of trypatse+chymase− and tryptase+chymase+ mast cells. IL-33 stimulation also increased the gene expression levels of TNF-α, IL-1β, IL-6, IL-17, RANKL, and MMP-9 in mast cells. Furthermore, IL-33 stimulated human CD14+ monocytes to differentiate into TRAP+ multinucleated osteoclasts. When CD14+ monocytes were co-cultured with mast cells, osteoclast differentiation was increased. Additionally, IL-33-activated mast cells stimulated osteoclast differentiation. The inhibition of intercellular contact between mast cells and monocytes using inserts reduced osteoclast differentiation. Conclusions IL-33 increased inflammatory and tissue-destructive cytokines by activation of mast cells. Mast cells stimulated osteoclast differentiation in monocytes. Mast cells could stimulate osteoclastogenesis indirectly through production of tissue-destructive cytokines and directly through stimulation of osteoclast precursors.
Collapse
Affiliation(s)
| | - Bo-Mi Kim
- Laboratory of Stem Cell, NEXEL, Seoul, South Korea
| | - Ji-Yeon Won
- R&D Center, OncoInsight Co. Ltd, Seoul, South Korea
| | - Hong-Ki Min
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, 05030, South Korea
| | - Kyung-Ann Lee
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, South Korea
| | - Sang-Heon Lee
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, 05030, South Korea
| | - Hae-Rim Kim
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul, 05030, South Korea.
| |
Collapse
|
15
|
Higher PGD 2 production by synovial mast cells from rheumatoid arthritis patients compared with osteoarthritis patients via miR-199a-3p/prostaglandin synthetase 2 axis. Sci Rep 2021; 11:5738. [PMID: 33707464 PMCID: PMC7952410 DOI: 10.1038/s41598-021-84963-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/22/2021] [Indexed: 12/29/2022] Open
Abstract
We previously reported that synovial mast cells (MCs) from patients with rheumatoid arthritis (RA) produced TNF-α in response to immune complexes via FcγRI and FcγRIIA. However, the specific functions of synovial MCs in RA remain unclear. This study aimed to elucidate those functions. Synovial tissues and fluid were obtained from RA and osteoarthritis (OA) patients undergoing joint replacement surgery. Synovium-derived, cultured MCs were generated by culturing dispersed synovial cells with stem cell factor. We performed microarray-based screening of mRNA and microRNA (miRNA), followed by quantitative RT-PCR-based verification. Synovial MCs from RA patients showed significantly higher prostaglandin systhetase (PTGS)1 and PTGS2 expression compared with OA patients' MCs, and they produced significantly more prostaglandin D2 (PGD2) following aggregation of FcγRI. PGD2 induced IL-8 production by human group 2 innate lymphoid cells, suggesting that PGD2-producing MCs induce neutrophil recruitment into the synovium of RA patients. PTGS2 mRNA expression in RA patients' MCs correlated inversely with miRNA-199a-3p expression, which down-regulated PTGS2. RA patients' synovial fluid contained significantly more PGD2 compared with OA patients' fluid. Synovial MCs might regulate inflammation in RA through hyper-production of PGD2 following FcRγ aggregation. Our findings indicate functional heterogeneity of human MCs among diseases.
Collapse
|
16
|
Toyoshima S, Sakamoto-Sasaki T, Kurosawa Y, Hayama K, Matsuda A, Watanabe Y, Terui T, Gon Y, Matsumoto K, Okayama Y. miR103a-3p in extracellular vesicles from FcεRI-aggregated human mast cells enhances IL-5 production by group 2 innate lymphoid cells. J Allergy Clin Immunol 2021; 147:1878-1891. [PMID: 33465368 DOI: 10.1016/j.jaci.2021.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/27/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Mast cells (MCs) are key regulators of IgE-mediated allergic inflammation. Cell-derived extracellular vesicles (EVs) contain bioactive compounds such as microRNAs. EVs can transfer signals to recipient cells, thus using a novel mechanism of cell-to-cell communication. However, whether MC-derived EVs are involved in FcεRI-mediated allergic inflammation is unclear. OBJECTIVE We sought to investigate the effect of EVs derived from FcεRI-aggregated human MCs on the function of human group 2 innate lymphoid cells (ILC2s). METHODS Human cultured MCs were sensitized with and without IgE for 1 hour and then incubated with anti-IgE antibody, IL-33, or medium alone for 24 hours. EVs in the MC supernatant were isolated by using ExoQuick-TC. RESULTS Coculture of ILC2s with EVs derived from the FcεRI-aggregated MCs significantly enhanced IL-5 production and sustained upregulation of IL-5 mRNA expression in IL-33-stimulated ILC2s, but IL-13 production and IL-13 mRNA expression were unchanged. miR103a-3p expression was upregulated in IL-33-stimulated ILC2s that had been cocultured with EVs derived from anti-IgE antibody-stimulated MCs. Transduction of an miR103a-3p mimic to ILC2s significantly enhanced IL-5 production by IL-33-stimulated ILC2s. miR103a-3p promoted demethylation of an arginine residue of GATA3 by downregulating protein arginine methyltransferase 5 (PRMT5) mRNA. Reduction of protein arginine methyltransferase 5 expression in ILC2s by using a small interfering RNA technique resulted in upregulation of IL-5 production by IL-33-stimulated ILC2s. Furthermore, the level of miR103a-3p expression was significantly higher in EVs from sera of patients with atopic dermatitis than in EVs from nonatopic healthy control subjects. CONCLUSION Eosinophilic allergic inflammation may be exacerbated owing to ILC2 activation by MC-derived miR103a-3p.
Collapse
Affiliation(s)
- Shota Toyoshima
- Allergy and Immunology Research Project Team, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan; Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan; Center for Medical Education, Nihon University School of Medicine, Tokyo, Japan
| | - Tomomi Sakamoto-Sasaki
- Allergy and Immunology Research Project Team, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan; Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan; Center for Medical Education, Nihon University School of Medicine, Tokyo, Japan
| | - Yusuke Kurosawa
- Allergy and Immunology Research Project Team, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan; Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan; Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Koremasa Hayama
- Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan; Divison of Cutaneous Science, Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Akira Matsuda
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasuo Watanabe
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tadashi Terui
- Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan; Divison of Cutaneous Science, Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuhiro Gon
- Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan; Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yoshimichi Okayama
- Allergy and Immunology Research Project Team, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan; Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan; Center for Medical Education, Nihon University School of Medicine, Tokyo, Japan.
| |
Collapse
|
17
|
Bhuiyan P, Wang YW, Sha HH, Dong HQ, Qian YN. Neuroimmune connections between corticotropin-releasing hormone and mast cells: novel strategies for the treatment of neurodegenerative diseases. Neural Regen Res 2021; 16:2184-2197. [PMID: 33818491 PMCID: PMC8354134 DOI: 10.4103/1673-5374.310608] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Corticotropin-releasing hormone is a critical component of the hypothalamic–pituitary–adrenal axis, which plays a major role in the body’s immune response to stress. Mast cells are both sensors and effectors in the interaction between the nervous and immune systems. As first responders to stress, mast cells can initiate, amplify and prolong neuroimmune responses upon activation. Corticotropin-releasing hormone plays a pivotal role in triggering stress responses and related diseases by acting on its receptors in mast cells. Corticotropin-releasing hormone can stimulate mast cell activation, influence the activation of immune cells by peripheral nerves and modulate neuroimmune interactions. The latest evidence shows that the release of corticotropin-releasing hormone induces the degranulation of mast cells under stress conditions, leading to disruption of the blood-brain barrier, which plays an important role in neurological diseases, such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, autism spectrum disorder and amyotrophic lateral sclerosis. Recent studies suggest that stress increases intestinal permeability and disrupts the blood-brain barrier through corticotropin-releasing hormone-mediated activation of mast cells, providing new insight into the complex interplay between the brain and gastrointestinal tract. The neuroimmune target of mast cells is the site at which the corticotropin-releasing hormone directly participates in the inflammatory responses of nerve terminals. In this review, we focus on the neuroimmune connections between corticotropin-releasing hormone and mast cells, with the aim of providing novel potential therapeutic targets for inflammatory, autoimmune and nervous system diseases.
Collapse
Affiliation(s)
- Piplu Bhuiyan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yi-Wei Wang
- Department of Anesthesiology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Huan-Huan Sha
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hong-Quan Dong
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yan-Ning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
18
|
Valent P, Akin C, Hartmann K, Nilsson G, Reiter A, Hermine O, Sotlar K, Sperr WR, Escribano L, George TI, Kluin-Nelemans HC, Ustun C, Triggiani M, Brockow K, Gotlib J, Orfao A, Kovanen PT, Hadzijusufovic E, Sadovnik I, Horny HP, Arock M, Schwartz LB, Austen KF, Metcalfe DD, Galli SJ. Mast cells as a unique hematopoietic lineage and cell system: From Paul Ehrlich's visions to precision medicine concepts. Am J Cancer Res 2020; 10:10743-10768. [PMID: 32929378 PMCID: PMC7482799 DOI: 10.7150/thno.46719] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
The origin and functions of mast cells (MCs) have been debated since their description by Paul Ehrlich in 1879. MCs have long been considered 'reactive bystanders' and 'amplifiers' in inflammatory processes, allergic reactions, and host responses to infectious diseases. However, knowledge about the origin, phenotypes and functions of MCs has increased substantially over the past 50 years. MCs are now known to be derived from multipotent hematopoietic progenitors, which, through a process of differentiation and maturation, form a unique hematopoietic lineage residing in multiple organs. In particular, MCs are distinguishable from basophils and other hematopoietic cells by their unique phenotype, origin(s), and spectrum of functions, both in innate and adaptive immune responses and in other settings. The concept of a unique MC lineage is further supported by the development of a distinct group of neoplasms, collectively referred to as mastocytosis, in which MC precursors expand as clonal cells. The clinical consequences of the expansion and/or activation of MCs are best established in mastocytosis and in allergic inflammation. However, MCs have also been implicated as important participants in a number of additional pathologic conditions and physiological processes. In this article, we review concepts regarding MC development, factors controlling MC expansion and activation, and some of the fundamental roles MCs may play in both health and disease. We also discuss new concepts for suppressing MC expansion and/or activation using molecularly-targeted drugs.
Collapse
|
19
|
Ragipoglu D, Dudeck A, Haffner-Luntzer M, Voss M, Kroner J, Ignatius A, Fischer V. The Role of Mast Cells in Bone Metabolism and Bone Disorders. Front Immunol 2020; 11:163. [PMID: 32117297 PMCID: PMC7025484 DOI: 10.3389/fimmu.2020.00163] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
Mast cells (MCs) are important sensor and effector cells of the immune system that are involved in many physiological and pathological conditions. Increasing evidence suggests that they also play an important role in bone metabolism and bone disorders. MCs are located in the bone marrow and secrete a wide spectrum of mediators, which can be rapidly released upon activation of mature MCs following their differentiation in mucosal or connective tissues. Many of these mediators can exert osteocatabolic effects by promoting osteoclast formation [e.g., histamine, tumor necrosis factor (TNF), interleukin-6 (IL-6)] and/or by inhibiting osteoblast activity (e.g., IL-1, TNF). By contrast, MCs could potentially act in an osteoprotective manner by stimulating osteoblasts (e.g., transforming growth factor-β) or reducing osteoclastogenesis (e.g., IL-12, interferon-γ). Experimental studies investigating MC functions in physiological bone turnover using MC-deficient mouse lines give contradictory results, reporting delayed or increased bone turnover or no influence depending on the mouse model used. By contrast, the involvement of MCs in various pathological conditions affecting bone is evident. MCs may contribute to the pathogenesis of primary and secondary osteoporosis as well as inflammatory disorders, including rheumatoid arthritis and osteoarthritis, because increased numbers of MCs were found in patients suffering from these diseases. The clinical observations could be largely confirmed in experimental studies using MC-deficient mouse models, which also provide mechanistic insights. MCs also regulate bone healing after fracture by influencing the inflammatory response toward the fracture, vascularization, bone formation, and callus remodeling by osteoclasts. This review summarizes the current view and understanding of the role of MCs on bone in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Deniz Ragipoglu
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Melanie Haffner-Luntzer
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Martin Voss
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jochen Kroner
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Anita Ignatius
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Trauma Research Center Ulm, Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
20
|
Min HK, Kim KW, Lee SH, Kim HR. Roles of mast cells in rheumatoid arthritis. Korean J Intern Med 2020; 35:12-24. [PMID: 31722515 PMCID: PMC6960056 DOI: 10.3904/kjim.2019.271] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory arthritis, and the complex interaction and activation of innate and adaptive immune cells are involved in RA pathogenesis. Mast cells (MCs) are one of the tissue-resident innate immune cells, and they contribute to RA pathogenesis. In the present review, the evidence of the pathologic role of MC in RA is discussed based on human and animal data. In addition, the potential role of MC in RA pathogenesis and the research area that should be focused on in the future are suggested.
Collapse
Affiliation(s)
- Hong Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, Seoul, Korea
| | - Kyoung-Woon Kim
- Conversant Research Consortium in Immunologic Disease, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sang-Heon Lee
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - Hae-Rim Kim
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
- Correspondence to Hae-Rim Kim, M.D. Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea Tel: +82-2-2030-7542, Fax: +82-2-2030-7728, E-mail:
| |
Collapse
|
21
|
Varricchi G, Pecoraro A, Loffredo S, Poto R, Rivellese F, Genovese A, Marone G, Spadaro G. Heterogeneity of Human Mast Cells With Respect to MRGPRX2 Receptor Expression and Function. Front Cell Neurosci 2019; 13:299. [PMID: 31333418 PMCID: PMC6616107 DOI: 10.3389/fncel.2019.00299] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022] Open
Abstract
Mast cells and their mediators play a role in the control of homeostasis and in the pathogenesis of several disorders. The concept of rodent mast cell heterogeneity, initially established in the mid-1960s has been extended in humans. Human mast cells isolated and purified from different anatomic sites can be activated via aggregation of cell surface high affinity IgE receptors (FcεRI) by antigens, superantigens, anti-IgE, and anti-FcεRI. MAS-related G protein-coupled receptor-X2 (MRGPRX2) is expressed at high level in human skin mast cells (MCs) (HSMCs), synovial MCs (HSyMCs), but not in lung MCs (HLMCs). MRGPX2 can be activated by neuropeptide substance P, several opioids, cationic drugs, and 48/80. Substance P (5 × 10−7 M – 5 × 10−6 M) induced histamine and tryptase release from HSMCs and to a lesser extent from HSyMCs, but not from HLMCs and human cardiac MCs (HHMCs). Morphine (10−5 M – 3 × 10−4 M) selectively induced histamine and tryptase release from HSMCs, but not from HLMCs and HHMCs. SP and morphine were incomplete secretagogues because they did not induce the de novo synthesis of arachidonic acid metabolites from human mast cells. In the same experiments anti-IgE (3 μg/ml) induced the release of histamine and tryptase and the de novo synthesis of prostaglandin D2 (PGD2) from HLMCs, HHMCs, HSyMCs, and HSMCs. By contrast, anti-IgE induced the production of leukotriene C4 (LTC4) from HLMCs, HHMCs, HSyMCs, but not from HSMCs. These results are compatible with the heterogeneous expression and function of MRGPRX2 receptor on primary human mast cells isolated from different anatomic sites.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization Center of Excellence, University of Naples Federico II, Naples, Italy
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization Center of Excellence, University of Naples Federico II, Naples, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Felice Rivellese
- Center for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Arturo Genovese
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization Center of Excellence, University of Naples Federico II, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization Center of Excellence, University of Naples Federico II, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council (CNR), Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,World Allergy Organization Center of Excellence, University of Naples Federico II, Naples, Italy
| |
Collapse
|
22
|
Wang Y, Nakahashi-Oda C, Okayama Y, Shibuya A. Autonomous regulation of IgE-mediated mast cell degranulation and immediate hypersensitivity reaction by an inhibitory receptor CD300a. J Allergy Clin Immunol 2019; 144:323-327.e7. [PMID: 31155312 DOI: 10.1016/j.jaci.2019.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/24/2019] [Accepted: 03/06/2019] [Indexed: 02/01/2023]
Affiliation(s)
- Yaqiu Wang
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; Life Science Center of Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Chigusa Nakahashi-Oda
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Yoshimichi Okayama
- Allergy and Immunology Project Team, Center for Institutional Research and Medical Education, Nihon University School of Medicine, Itabashi-Ku, Tokyo, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; Life Science Center of Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
23
|
Wang Q, Lepus CM, Raghu H, Reber LL, Tsai MM, Wong HH, von Kaeppler E, Lingampalli N, Bloom MS, Hu N, Elliott EE, Oliviero F, Punzi L, Giori NJ, Goodman SB, Chu CR, Sokolove J, Fukuoka Y, Schwartz LB, Galli SJ, Robinson WH. IgE-mediated mast cell activation promotes inflammation and cartilage destruction in osteoarthritis. eLife 2019; 8:39905. [PMID: 31084709 PMCID: PMC6516833 DOI: 10.7554/elife.39905] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 04/10/2019] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis is characterized by articular cartilage breakdown, and emerging evidence suggests that dysregulated innate immunity is likely involved. Here, we performed proteomic, transcriptomic, and electron microscopic analyses to demonstrate that mast cells are aberrantly activated in human and murine osteoarthritic joint tissues. Using genetic models of mast cell deficiency, we demonstrate that lack of mast cells attenuates osteoarthritis in mice. Using genetic and pharmacologic approaches, we show that the IgE/FcεRI/Syk signaling axis is critical for the development of osteoarthritis. We find that mast cell-derived tryptase induces inflammation, chondrocyte apoptosis, and cartilage breakdown. Our findings demonstrate a central role for IgE-dependent mast cell activation in the pathogenesis of osteoarthritis, suggesting that targeting mast cells could provide therapeutic benefit in human osteoarthritis. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Qian Wang
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Christin M Lepus
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Harini Raghu
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Laurent L Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, United States
| | - Mindy M Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, United States
| | - Heidi H Wong
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Ericka von Kaeppler
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Nithya Lingampalli
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Michelle S Bloom
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Nick Hu
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Eileen E Elliott
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Francesca Oliviero
- Rheumatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Leonardo Punzi
- Rheumatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Nicholas J Giori
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, United States
| | - Stuart B Goodman
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, United States
| | - Constance R Chu
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, United States
| | - Jeremy Sokolove
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| | - Yoshihiro Fukuoka
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, United States
| | - Lawrence B Schwartz
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, United States
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, United States.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, United States
| | - William H Robinson
- GRECC, VA Palo Alto Health Care System, Palo Alto, United States.,Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
24
|
Rivellese F, Rossi FW, Galdiero MR, Pitzalis C, de Paulis A. Mast Cells in Early Rheumatoid Arthritis. Int J Mol Sci 2019; 20:ijms20082040. [PMID: 31027208 PMCID: PMC6515166 DOI: 10.3390/ijms20082040] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 04/18/2019] [Accepted: 04/22/2019] [Indexed: 12/17/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by inflammation of the synovial membrane, with thickening of the synovial layer, cellular hyperplasia, and infiltration of immune cells. Mast cells (MCs) are cells of the innate immunity present in healthy synovia and part of the cellular hyperplasia characterizing RA synovitis. Although their presence in synovia has been well described, the exact functions and the correlation of MCs with disease development and progression have been debated, particularly because of contradictory data obtained in animal models and from patients with longstanding disease. Here, we present a revision of the literature on MCs in RA, including the most recent observations obtained from patients with early RA, indicating MCs as relevant markers of disease severity in early RA.
Collapse
Affiliation(s)
- Felice Rivellese
- William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Francesca Wanda Rossi
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
| | - Costantino Pitzalis
- William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| | - Amato de Paulis
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy.
| |
Collapse
|
25
|
Harshan S, Dey P, Ragunathan S. Effects of rheumatoid arthritis associated transcriptional changes on osteoclast differentiation network in the synovium. PeerJ 2018; 6:e5743. [PMID: 30324023 PMCID: PMC6186409 DOI: 10.7717/peerj.5743] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/12/2018] [Indexed: 12/15/2022] Open
Abstract
Background Osteoclast differentiation in the inflamed synovium of rheumatoid arthritis (RA) affected joints leads to the formation of bone lesions. Reconstruction and analysis of protein interaction networks underlying specific disease phenotypes are essential for designing therapeutic interventions. In this study, we have created a network that captures signal flow leading to osteoclast differentiation. Based on transcriptome analysis, we have indicated the potential mechanisms responsible for the phenotype in the RA affected synovium. Method We collected information on gene expression, pathways and protein interactions related to RA from literature and databases namely Gene Expression Omnibus, Kyoto Encyclopedia of Genes and Genomes pathway and STRING. Based on these information, we created a network for the differentiation of osteoclasts. We identified the differentially regulated network genes and reported the signaling that are responsible for the process in the RA affected synovium. Result Our network reveals the mechanisms underlying the activation of the neutrophil cytosolic factor complex in connection to osteoclastogenesis in RA. Additionally, the study reports the predominance of the canonical pathway of NF-κB activation in the diseased synovium. The network also confirms that the upregulation of T cell receptor signaling and downregulation of transforming growth factor beta signaling pathway favor osteoclastogenesis in RA. To the best of our knowledge, this is the first comprehensive protein–protein interaction network describing RA driven osteoclastogenesis in the synovium. Discussion This study provides information that can be used to build models of the signal flow involved in the process of osteoclast differentiation. The models can further be used to design therapies to ameliorate bone destruction in the RA affected joints.
Collapse
Affiliation(s)
- Shilpa Harshan
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
| | - Poulami Dey
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India.,Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Srivatsan Ragunathan
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, Karnataka, India
| |
Collapse
|
26
|
Matsuno T, Toyoshima S, Sakamoto-Sasaki T, Kashiwakura JI, Matsuda A, Watanabe Y, Azuma H, Kawana K, Yamamoto T, Okayama Y. Characterization of human decidual mast cells and establishment of a culture system. Allergol Int 2018; 67S:S18-S24. [PMID: 29784282 DOI: 10.1016/j.alit.2018.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/12/2018] [Accepted: 04/25/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Although rodent decidual mast cells (MCs) reportedly play an important role in implantation and placenta formation, the characterization of human decidual MCs has been not well clarified. The aims of this study were to investigate the distribution and characteristics of MCs in human decidua and to establish a culture system for decidua-derived MCs. METHODS Decidual tissues were obtained from patients who underwent a legal elective abortion (6th week to 9th week of pregnancy), and decidual MCs were enzymatically dispersed. Cultured decidua-derived MCs were generated by culturing decidual cells with stem cell factor. An ultrastructural analysis of primary decidual MCs and cultured decidua-derived MCs was performed using a transmission electron microscope. Receptor and protease expression was analyzed using FACS. Histamine released from MCs was measured using enzyme immune assays. RESULTS A larger proportion of tryptase positive(+) MCs in decidua was present on the maternal side. Both enzymatically dispersed decidual MCs and cultured decidua-derived MCs showed an FcεRIα+Kit+tryptase+chymase+ phenotype. Their granules contenting particles exhibited variable amounts of electron-lucent space separating electron-dense particles. Both enzymatically dispersed decidual MCs and cultured decidua-derived MCs released comparable amounts of histamine following FcεRI aggregation. CONCLUSIONS The isolation method for MCs from decidua during early pregnancy and the culture system for decidua-derived MCs may enable the roles of decidual MC during pregnancy to be explored.
Collapse
Affiliation(s)
- Takayuki Matsuno
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Tokyo, Japan; Allergy and Immunology Project Team, Nihon University School of Medicine, Tokyo, Japan
| | - Shota Toyoshima
- Allergy and Immunology Project Team, Nihon University School of Medicine, Tokyo, Japan; Center for Institutional Research and Medical Education, Nihon University School of Medicine, Tokyo, Japan
| | - Tomomi Sakamoto-Sasaki
- Allergy and Immunology Project Team, Nihon University School of Medicine, Tokyo, Japan; Center for Institutional Research and Medical Education, Nihon University School of Medicine, Tokyo, Japan
| | - Jun-Ichi Kashiwakura
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Akira Matsuda
- Department of Ophthalmology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yasuo Watanabe
- Department of Ophthalmology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiromitsu Azuma
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Tokyo, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Nihon University School of Medicine, Tokyo, Japan
| | - Tatsuo Yamamoto
- Department of Obstetrics and Gynecology, Kasukabe Medical Center, Saitama, Japan
| | - Yoshimichi Okayama
- Allergy and Immunology Project Team, Nihon University School of Medicine, Tokyo, Japan; Center for Institutional Research and Medical Education, Nihon University School of Medicine, Tokyo, Japan.
| |
Collapse
|
27
|
Shimanaka Y, Kono N, Taketomi Y, Arita M, Okayama Y, Tanaka Y, Nishito Y, Mochizuki T, Kusuhara H, Adibekian A, Cravatt BF, Murakami M, Arai H. Omega-3 fatty acid epoxides are autocrine mediators that control the magnitude of IgE-mediated mast cell activation. Nat Med 2017; 23:1287-1297. [DOI: 10.1038/nm.4417] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 09/07/2017] [Indexed: 02/08/2023]
|
28
|
|
29
|
Okamura Y, Mishima S, Kashiwakura JI, Sasaki-Sakamoto T, Toyoshima S, Kuroda K, Saito S, Tokuhashi Y, Okayama Y. The dual regulation of substance P-mediated inflammation via human synovial mast cells in rheumatoid arthritis. Allergol Int 2017; 66S:S9-S20. [PMID: 28366675 DOI: 10.1016/j.alit.2017.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/13/2017] [Accepted: 02/27/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Neural pathways are thought to be directly involved in the pathogenesis of rheumatoid arthritis (RA). Although synovial mast cells (MCs) are activated by substance P (SP), the role of MCs in neural pathways in RA remains unknown. The aims of this study were to investigate 1) whether tachykinins are produced by synovial MCs and whether production differs in RA and osteoarthritis (OA) patients, and 2) what is the responsible receptor for SP in synovial MCs. METHODS Synovial tissues were obtained from patients with RA or OA undergoing joint replacement surgery. Cultured synovium-derived MCs were generated by culturing dispersed synovial cells with stem cell factor. SP expression was investigated using immunofluorescence and enzyme immunoassays. Mas-related gene X2 (MrgX2) expression was reduced in human MCs using a lentiviral shRNA silencing technique. RESULTS SP expression was localized around the cell membrane in 41% (median) of the MCs in synovium from RA but in only 7% of that from OA, suggesting the activation of MCs. Synovial MCs expressed tachykinin (TAC) 1 mRNA, the expression of which was upregulated by the aggregation of FcɛRI or the addition of aggregated IgG. However, the released SP appeared to be rapidly degraded by MC chymase. Synovial MCs were activated with SP through MrgX2 to release histamine without producing proinflammatory cytokines. CONCLUSIONS Activated synovial MCs may rapidly degrade SP, which may downregulate the SP-mediated activation of synoviocytes in RA. On the other hand, SP activates MCs to induce inflammatory mediators, suggesting the dual regulation of SP-mediated inflammation by MCs in RA.
Collapse
|
30
|
Ali H. Emerging Roles for MAS-Related G Protein-Coupled Receptor-X2 in Host Defense Peptide, Opioid, and Neuropeptide-Mediated Inflammatory Reactions. Adv Immunol 2017; 136:123-162. [PMID: 28950944 DOI: 10.1016/bs.ai.2017.06.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mast cells (MCs) are tissue-resident immune cells that contribute to host defense but are best known for their roles in allergic and inflammatory diseases. In humans, MCs are divided into two subtypes based on the protease content of their secretory granules. Thus, human lung MCs contain only tryptase and are known as MCT, whereas skin MCs contain both tryptase and chymase and are known as MCTC. Patients with severe asthma display elevated MCs in the lung, which undergo phenotypic change from MCT to MCTC. Although the human genome contains four Mas related G protein coupled receptor X (MRGPRX) genes, an important feature of MCTC is that they selectively express MRGPRX2. It is activated by antimicrobial host defense peptides such as human β-defensins and the cathelicidin LL-37 and likely contributes to host defense. MRGPRX2 is also a receptor for the neuropeptide substance P, major basic protein, eosinophil peroxidase, opioids, and many FDA-approved cationic drugs. Increased expression of MRGPRX2 or enhanced downstream signaling likely contributes to chronic inflammatory diseases such as rosacea, atopic dermatitis, chronic urticaria, and severe asthma. In this chapter, I will discuss the expression profile and function of MRGPRX1-4 and review the emerging roles of MRGPRX2 on host defense, chronic inflammatory diseases, and drug-induced pseudoallergic reactions. I will also examine the novel aspects of MRGPRX2 signaling in MCs as it related to degranulation and review the mechanisms of its regulation.
Collapse
Affiliation(s)
- Hydar Ali
- University of Pennsylvania School of Dental Medicine, Philadelphia, PA, United States.
| |
Collapse
|
31
|
Rivellese F, Nerviani A, Rossi FW, Marone G, Matucci-Cerinic M, de Paulis A, Pitzalis C. Mast cells in rheumatoid arthritis: friends or foes? Autoimmun Rev 2017; 16:557-563. [PMID: 28411167 DOI: 10.1016/j.autrev.2017.04.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 02/17/2017] [Indexed: 12/21/2022]
Abstract
Mast cells are tissue-resident cells of the innate immunity, implicated in the pathogenesis of many autoimmune diseases, including rheumatoid arthritis (RA). They are present in synovia and their activation has been linked to the potentiation of inflammation in the course of RA. However, recent investigations questioned the role of mast cells in arthritis. In particular, animal models generated conflicting results, so that many of their pro-inflammatory, i.e. pro-arthritogenic functions, even though supported by robust experimental evidence, have been labelled as redundant. At the same time, a growing body of evidence suggests that mast cells can act as tunable immunomodulatory cells. These characteristics, not yet fully understood in the context of RA, could partially explain the inconsistent results obtained with experimental models, which do not account for the pro- and anti-inflammatory functions exerted in more chronic heterogeneous conditions such as RA. Here we present an overview of the current knowledge on mast cell involvement in RA, including the intriguing hypothesis of mast cells acting as subtle immunomodulatory cells and the emerging concept of synovial mast cells as potential biomarkers for patient stratification.
Collapse
Affiliation(s)
- Felice Rivellese
- William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Alessandra Nerviani
- William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesca Wanda Rossi
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), Naples, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology AOUC, University of Florence, Florence, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Costantino Pitzalis
- William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
32
|
Kan JI, Mishima S, Kashiwakura JI, Sasaki-Sakamoto T, Seki M, Saito S, Ra C, Tokuhashi Y, Okayama Y. Interleukin-17A expression in human synovial mast cells in rheumatoid arthritis and osteoarthritis. Allergol Int 2016; 65 Suppl:S11-6. [PMID: 27209051 DOI: 10.1016/j.alit.2016.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/01/2016] [Accepted: 04/18/2016] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Interleukin (IL)-17A plays a pivotal role in the pathogenesis of rheumatoid arthritis (RA). The expression of IL-17A in synovial mast cells (MCs) in RA and osteoarthritis (OA) has been reported, but the frequencies of IL-17A expression in synovial MCs have varied. The aim of this study was to investigate whether IL-17A expression is upregulated in human synovial MCs in RA and to elucidate the mechanism of IL-17A expression in synovial MCs. METHODS Synovial tissues were obtained from patients with RA or OA undergoing joint replacement surgery, and synovial MCs were enzymatically dispersed. Synovium-derived cultured MCs were generated by culturing synovial cells with stem cell factor. IL-17A expression was investigated using immunofluorescence in synovial tissues. IL-17A mRNA expression and its production from MCs were examined using RT-PCR and ELISA, respectively. RESULTS The number of IL-17A-positive ((+)) synovial MCs and the percentage of IL-17A(+) MCs among all the IL-17A(+) cells from RA patients were not significantly increased compared with those from OA subjects. The synovium-derived cultured MCs spontaneously released small amounts of IL-17A. Neither IgE- nor IgG-dependent stimulation increased IL-17A production from the MCs. IL-33, tumor necrosis factor-α, C5a, lipopolysaccharide or IL-23 plus IL-1β did not affect IL-17A production in MCs. CONCLUSIONS The synovial MCs are not a main source of IL-17A in RA.
Collapse
|
33
|
James LK, Till SJ. Potential Mechanisms for IgG4 Inhibition of Immediate Hypersensitivity Reactions. Curr Allergy Asthma Rep 2016; 16:23. [PMID: 26892721 PMCID: PMC4759210 DOI: 10.1007/s11882-016-0600-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
IgG4 is the least abundant IgG subclass in human serum, representing less than 5 % of all IgG. Increases in IgG4 occur following chronic exposure to antigen and are generally associated with states of immune tolerance. In line with this, IgG4 is regarded as an anti-inflammatory antibody with a limited ability to elicit effective immune responses. Furthermore, IgG4 attenuates allergic responses by inhibiting the activity of IgE. The mechanism by which IgG4 inhibits IgE-mediated hypersensitivity has been investigated using a variety of model systems leading to two proposed mechanisms. First by sequestering antigen, IgG4 can function as a blocking antibody, preventing cross-linking of receptor bound IgE. Second IgG4 has been proposed to co-stimulate the inhibitory IgG receptor FcγRIIb, which can negatively regulate FcεRI signaling and in turn inhibit effector cell activation. Recent advances in our understanding of the structural features of human IgG4 have shed light on the unique functional and immunologic properties of IgG4. The aim of this review is to evaluate our current understanding of IgG4 biology and reassess the mechanisms by which IgG4 functions to inhibit IgE-mediated allergic responses.
Collapse
Affiliation(s)
- Louisa K James
- Randall Division of Cell and Molecular Biophysics and MRC and Asthma UK Centre for Allergic Mechanisms of Asthma, King's College London, London, SE1 1UL, UK.
| | - Stephen J Till
- Division of Asthma, Allergy and Lung Biology, King's College London and Department of Allergy, Guy's and St. Thomas' NHS Foundation Trust, London, SE1 9RT, UK.
| |
Collapse
|
34
|
Lopes F, Graepel R, Reyes JL, Wang A, Petri B, McDougall JJ, Sharkey KA, McKay DM. Involvement of Mast Cells in α7 Nicotinic Receptor Agonist Exacerbation of Freund's Complete Adjuvant-Induced Monoarthritis in Mice. Arthritis Rheumatol 2016; 68:542-52. [PMID: 26314943 DOI: 10.1002/art.39411] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 08/25/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Activation of antiinflammatory cholinergic (vagal) pathways can reduce inflammation, and in vitro studies support a pivotal role of α7 nicotinic acetylcholine receptors (α7-nAChR), macrophages, and T cells in these events. The aim of this study was to assess α7-nAChR agonists as an antiinflammatory treatment for Freund's complete adjuvant (CFA)-induced monoarthritis. METHODS Arthritis was induced by intraarticular injection of CFA unilaterally into the knee joints of mice. Animals were treated with α7-nAChR agonists (AR-R17779 or A844606), with or without antagonists (COG133 or methyllycaconitine), and joint inflammation and pain were assessed. Experiments were repeated in c-Kit(W-sh) mast cell-deficient mice, and the effects of an α7-nAChR agonist on mast cell proliferation, migration, and activation by lipopolysaccharide (LPS) were tested. RESULTS Treatment with α7-nAChR agonists significantly exacerbated CFA-induced arthritis and pain, as gauged by all indices of assessment, the specificity of which was confirmed by coadministration of an nAChR antagonist that attenuated the increase in disease severity. Toluidine blue-positive mast cells were increased in the joint capsule of CFA plus AR-R17779-treated mice, and AR-R17779 enhanced LPS-induced TNF proliferation and migration of a human mast cell line. The AR-R17779-driven increase in severity of CFA-induced arthritis was significantly reduced in mast cell-deficient mice. CONCLUSION Using CFA to elicit a local inflammatory response, we found that pharmacologic activation of α7-nAChR exacerbated joint inflammation and pain, in part via mast cells, which illustrates the organ- and disease-specific nature of regulatory neuroimmune mechanisms. Thus, α7-nAChR activation may not be uniformly antiinflammatory in all types of inflammatory joint disease.
Collapse
Affiliation(s)
| | | | | | - Arthur Wang
- University of Calgary, Calgary, Alberta, Canada
| | - Björn Petri
- University of Calgary, Calgary, Alberta, Canada
| | | | | | | |
Collapse
|
35
|
Suurmond J, van der Velden D, Kuiper J, Bot I, Toes RE. Mast cells in rheumatic disease. Eur J Pharmacol 2016; 778:116-24. [DOI: 10.1016/j.ejphar.2015.03.085] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/13/2015] [Accepted: 03/25/2015] [Indexed: 12/18/2022]
|
36
|
Cho C, Nguyen A, Bryant KJ, O'Neill SG, McNeil HP. Prostaglandin D2 metabolites as a biomarker of in vivo mast cell activation in systemic mastocytosis and rheumatoid arthritis. IMMUNITY INFLAMMATION AND DISEASE 2015; 4:64-9. [PMID: 27042302 PMCID: PMC4768064 DOI: 10.1002/iid3.94] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/10/2015] [Accepted: 11/11/2015] [Indexed: 12/22/2022]
Abstract
Mast cells (MCs) participate in diseases such as systemic mastocytosis (SM) and allergic conditions. Less well understood is the role of MCs in non‐allergic inflammatory disorders like rheumatoid arthritis (RA). Studying definitive roles for MCs in human diseases has been hampered by the lack of a well‐accepted biomarker for monitoring in vivo MC activation. This study aimed to investigate the utility of urinary tetranor PGDM (T‐PGDM) as a biomarker of in vivo MC activation in patients with SM, and apply this biomarker to assess MC involvement in relation to RA disease activity. A prospective, cross‐sectional cohort study was conducted to measure a major urinary metabolite of prostaglandin D2, T‐PGDM. Urine samples were collected from patients with RA (n = 60), SM (n = 17) and healthy normal controls (n = 16) and T‐PGDM excretion was determined by enzyme immunoassay as nanograms per milligram of urinary creatinine (ng/mg Cr). Mean urinary T‐PGDM excretion was significantly higher (p < 0.01) in patients with SM compared to controls (37.2 vs. 11.5 ng/mg Cr) with 65% of SM patients showing elevated levels. One third of patients with RA had elevated T‐PGDM excretion, and the mean level in the RA group (20.0 ng/mg Cr) was significantly higher than controls (p < 0.01). Medications inhibiting cyclooxygenase reduced T‐PGDM excretion. Urinary T‐PGDM excretion appears promising as a biomarker of in vivo MC activity and elevated levels in 33% of patients with RA provides evidence of MC activation in this disease.
Collapse
Affiliation(s)
- Catherine Cho
- Faculty of Medicine University of New South Wales Sydney 2052 Australia
| | - Anna Nguyen
- Faculty of Medicine University of New South Wales Sydney 2052 Australia
| | | | - Sean G O'Neill
- Faculty of Medicine University of New South Wales Sydney 2052 Australia
| | - H Patrick McNeil
- Faculty of Medicine and Health Sciences Macquarie University Sydney 2109 Australia
| |
Collapse
|
37
|
Nunomura S, Okayama Y, Matsumoto K, Hashimoto N, Endo-Umeda K, Terui T, Makishima M, Ra C. Activation of LXRs using the synthetic agonist GW3965 represses the production of pro-inflammatory cytokines by murine mast cells. Allergol Int 2015; 64 Suppl:S11-7. [PMID: 26344074 DOI: 10.1016/j.alit.2015.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/26/2015] [Accepted: 03/03/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The activation of liver X receptor (LXR) α or LXRβ negatively regulates the expression of pro-inflammatory genes in mammalian cells. We recently reported that 25-hydroxycholesterol, a representative LXR-activating oxysterol, suppresses IL-6 production in mouse mast cells (MCs) following its engagement of the high-affinity IgE receptor (FcεRI). This finding suggests that murine MCs express functional LXRs; however, the mechanisms underlying the LXR-dependent repression of the MC-mediated production of pro-inflammatory cytokines, including IL-6, are poorly understood. Therefore, we employed the synthetic LXR ligand GW3965 to examine the functions of LXRα and LXRβ in the production of pro-inflammatory cytokines by murine bone marrow-derived MCs (BMMCs). METHODS We prepared BMMCs from wild-type (WT), LXRα(-/-), and LXRα/β(-/-) mice. Each group of BMMCs was pretreated with GW3965 and then stimulated with IgE+antigen (Ag) or lipopolysaccharide (LPS). Cytokine production was then analyzed using specific ELISA kits. RESULTS The activation of LXRs by GW3965 significantly attenuated the production of IL-1α and IL-1β, but not of IL-6, in the WT and LXRα(-/-) BMMCs stimulated with IgE+Ag. However, GW3965 treatment decreased the production of IL-1α, IL-1β, and IL-6 in WT and LXRα(-/-) BMMCs upon stimulation with LPS, while the GW3965-mediated suppression of cytokine production was nearly absent from the LXRα/β(-/-) BMMCs. CONCLUSIONS These findings demonstrate, for the first time, that the activation of LXRs by GW3965 attenuates the antigen- or LPS-induced production of pro-inflammatory cytokines, such as IL-1α and IL-1β, in murine MCs and that LXRβ plays an important role in the LXR-mediated repression of cytokine production.
Collapse
|
38
|
Reber LL, Sibilano R, Mukai K, Galli SJ. Potential effector and immunoregulatory functions of mast cells in mucosal immunity. Mucosal Immunol 2015; 8:444-63. [PMID: 25669149 PMCID: PMC4739802 DOI: 10.1038/mi.2014.131] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 11/27/2014] [Indexed: 02/04/2023]
Abstract
Mast cells (MCs) are cells of hematopoietic origin that normally reside in mucosal tissues, often near epithelial cells, glands, smooth muscle cells, and nerves. Best known for their contributions to pathology during IgE-associated disorders such as food allergy, asthma, and anaphylaxis, MCs are also thought to mediate IgE-associated effector functions during certain parasite infections. However, various MC populations also can be activated to express functional programs--such as secreting preformed and/or newly synthesized biologically active products--in response to encounters with products derived from diverse pathogens, other host cells (including leukocytes and structural cells), damaged tissue, or the activation of the complement or coagulation systems, as well as by signals derived from the external environment (including animal toxins, plant products, and physical agents). In this review, we will discuss evidence suggesting that MCs can perform diverse effector and immunoregulatory roles that contribute to homeostasis or pathology in mucosal tissues.
Collapse
Affiliation(s)
- Laurent L Reber
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| | - Riccardo Sibilano
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| | - Kaori Mukai
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| | - Stephen J Galli
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA,Department of Microbiology & Immunology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| |
Collapse
|
39
|
Sellge G, Barkowsky M, Kramer S, Gebhardt T, Sander LE, Lorentz A, Bischoff SC. Interferon-γ regulates growth and controls Fcγ receptor expression and activation in human intestinal mast cells. BMC Immunol 2014; 15:27. [PMID: 24996251 PMCID: PMC4227132 DOI: 10.1186/1471-2172-15-27] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 06/20/2014] [Indexed: 01/05/2023] Open
Abstract
Background Development and function of tissue resident mast cells (MCs) is tightly controlled by various cytokines, most of which belong to the typical T helper (Th) 2-type cytokines such as IL-3 and IL-4. The effects of the Th1-type cytokine IFN-γ on human MCs is less clear. Results Here, we analyzed the effects of IFN-γ on tissue-derived, mature human MCs. We found that INF-γ decreases proliferation, without affecting apoptosis in human intestinal MCs cultured in the presence of optimal concentrations of stem cell factor (SCF) or SCF and IL-4. However, in the absence of growth factors or at suboptimal concentrations of SCF, INF-γ promotes survival through inhibition of MC apoptosis. Interestingly, we found that INF-γ has no effect on FcϵRI expression and FcϵRI-mediated release of histamine and leukotriene (LT)C4, while it has profound effects on FcγR expression and activation. We show that intestinal MCs express FcγRI, FcγRIIa, and FcγRIIc, whereas FcγRIIb expression was found in only 40% of the isolates and FcγRIII was never detectable. INF-γ strongly increases FcγRI and decreases FcγRIIa expression. INF-γ-naïve MCs produce LTC4 but fail to degranulate upon crosslinking of surface-bound monomeric IgG. In contrast, INF-γ-treated MCs rapidly release granule-stored histamine in addition to de novo-synthesized LTC4. Conclusion In summary, we identify INF-γ as an important regulator of tissue-resident human MCs. IFN-γ displays a dual function by blocking extensive MC proliferation, while decreasing apoptosis in starving MCs and enhancing FcγRI expression and activation. These results emphasize the involvement of mucosal MCs in Th1-mediated disorders.
Collapse
Affiliation(s)
- Gernot Sellge
- Department of Internal Medicine III, University Hospital Aachen, RWTH University, Aachen, Germany.
| | | | | | | | | | | | | |
Collapse
|
40
|
Fujisawa D, Kashiwakura JI, Kita H, Kikukawa Y, Fujitani Y, Sasaki-Sakamoto T, Kuroda K, Nunomura S, Hayama K, Terui T, Ra C, Okayama Y. Expression of Mas-related gene X2 on mast cells is upregulated in the skin of patients with severe chronic urticaria. J Allergy Clin Immunol 2014; 134:622-633.e9. [PMID: 24954276 DOI: 10.1016/j.jaci.2014.05.004] [Citation(s) in RCA: 270] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 02/26/2014] [Accepted: 05/01/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND Wheal reactions to intradermally injected neuropeptides, such as substance P (SP) and vasoactive intestinal peptide, are significantly larger and longer lasting in patients with chronic urticaria (CU) than in nonatopic control (NC) subjects. Mas-related gene X2 (MrgX2) has been identified as a receptor for basic neuropeptides, such as SP and vasoactive intestinal peptide. Mast cell (MC) responsiveness to eosinophil mediators contributes to the late-phase reaction of allergy. OBJECTIVE We sought to compare the frequency of MrgX2 expression in skin MCs from patients with CU and NC subjects and to identify the receptor for basic eosinophil granule proteins on human skin MCs. METHODS MrgX2 expression was investigated by using immunofluorescence in skin tissues from NC subjects and patients with severe CU and on skin-derived cultured MCs. MrgX2 expression in human MCs was reduced by using a lentiviral small hairpin RNA silencing technique. Ca(2+) influx was measured in CHO cells transfected with MrgX2 in response to eosinophil granule proteins. Histamine and prostaglandin D2 levels were measured by using enzyme immunoassays. RESULTS The number of MrgX2(+) skin MCs and the percentage of MrgX2(+) MCs in all MCs in patients with CU were significantly greater than those in NC subjects. Eosinophil infiltration in urticarial lesions was observed in 7 of 9 patients with CU. SP, major basic protein, and eosinophil peroxidase, but not eosinophil-derived neurotoxin, induced histamine release from human skin MCs through MrgX2. CONCLUSION MrgX2 might be a new target molecule for the treatment of wheal reactions in patients with severe CU.
Collapse
Affiliation(s)
- Daisuke Fujisawa
- Allergy and Immunology Group, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan; Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Jun-Ichi Kashiwakura
- Laboratory for Allergic Disease, RCAI, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama, Japan
| | - Hirohito Kita
- Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minn
| | - Yusuke Kikukawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Fujisawa, Japan
| | - Yasushi Fujitani
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Fujisawa, Japan
| | - Tomomi Sasaki-Sakamoto
- Allergy and Immunology Group, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan
| | - Kazumichi Kuroda
- Department of Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Satoshi Nunomura
- Allergy and Immunology Group, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan; Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Koremasa Hayama
- Allergy and Immunology Group, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan; Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Tadashi Terui
- Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Chisei Ra
- Department of Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Yoshimichi Okayama
- Allergy and Immunology Group, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan.
| |
Collapse
|
41
|
Bader-Meunier B, Bulai Livideanu C, Larroche C, Durieu I, Artru L, Beucher A, Cormier G, Cornec D, Delarco M, Dubost JJ, Fontaine C, Gourin MP, Javier RM, de Jauréguiberry JP, Maisonneuve H, Toussirot E, Ugo V, Echaubard S, Mahlaoui N, Aouba A, Bodemer C, Briot K, Frenzel L, Lortholary O, Chandesris MO, Hermine O. Association of mastocytosis with inflammatory joint diseases: a series of 31 patients. Semin Arthritis Rheum 2014; 44:362-5. [PMID: 25037281 DOI: 10.1016/j.semarthrit.2014.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/10/2014] [Accepted: 05/12/2014] [Indexed: 01/29/2023]
Abstract
OBJECTIVES We studied the clinical phenotypes and tolerance to treatments in a series of patients affected by both inflammatory joint diseases and mastocytosis. METHODS This retrospective multicenter study was conducted on behalf of 3 networks focused on mastocytosis, pediatric, and adults' inflammatory joint diseases. Patients who displayed both mastocytosis and inflammatory joint diseases were included. RESULTS A total of 31 patients were included. They had spondyloarthritis (SpA) (16 patients), rheumatoid arthritis (6 patients), juvenile idiopathic arthritis (2 patients), and undifferentiated arthritis (7 patients). The median ages at diagnosis of arthritis and mastocytosis were 44 and 40.5 years, respectively. Disease-modifying anti-rheumatic drugs (DMARDs) were required in 22 patients, comprising mostly methotrexate (13 patients), salazopyrin (8 patients), anti-tumor-necrosis-factor agents (7 patients), and corticosteroids (9 patients). They were well tolerated. Adverse events occurred in 2/24 patients receiving non-steroidal anti-inflammatory drugs. The prevalence of SpA among the 600 patients included in the mastocytosis cohort was 2.33%, which is significantly higher than the prevalence of SpA in the French population (p < 0.001). CONCLUSIONS This study suggests that mastocytosis is associated with a higher prevalence of SpA than expected, and that DMARDs, notably anti-TNFα agents, are well tolerated in patients with mastocytosis. Mast cells might be involved in the development of SpA.
Collapse
Affiliation(s)
- Brigitte Bader-Meunier
- Department of Pediatric Immunology and Rheumatology, INSERM U768, Hôpital Necker Enfants Malades (AP-HP), Paris 75015, France; Institut Imagine, Université Sorbonne Paris cité, Hôpital Necker-Enfants Malades (AP-HP), Paris, France.
| | | | - Claire Larroche
- Department of Internal Medicine, CHU Avicenne, (AP-HP), Bobigny, France
| | - Isabelle Durieu
- Department of Internal Medicine, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Université de Lyon, Pierre Bénite, France
| | - Laure Artru
- Department of Rheumatology, Hôpital du Mans, Le Mans, France
| | - Anne Beucher
- Department of Internal Medicine, CHU Angers, Angers, France
| | - Grégoire Cormier
- Department of Rheumatology, CHD Vendée, La Roche sur Yon, France
| | - Divi Cornec
- Department of Rheumatology, CHRU Brest, Brest, France
| | - Michel Delarco
- Department of Rheumatology, Hôpital de Bigorre, Tarbes, France
| | - Jean-Jacques Dubost
- Department of Rheumatology, Hopital G. Montpied, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Christophe Fontaine
- Department of Internal Medicine, Clinique Mutualiste Beau Soleil, Montpellier, France
| | | | | | | | - Hervé Maisonneuve
- Department of Internal Medicine, CHD les Oudairies, La Roche sur Yon, France
| | - Eric Toussirot
- Clinical Investigation Center Biotherapy INSERM CBT-506, CHRU de Besançon, Besançon, France; Department of Rheumatology, CHRU de Besançon, Besançon, France
| | - Valérie Ugo
- Laboratory of Hematology, CHU de Brest, Brest, France
| | | | - Nizar Mahlaoui
- Department of Pediatric Immunology and Rheumatology, INSERM U768, Hôpital Necker Enfants Malades (AP-HP), Paris 75015, France
| | - Achille Aouba
- Department of Hematology, Hôpital Necker EnfantsMalades (AP-HP), Paris, France; Institut Imagine, Université Sorbonne Paris cité, Hôpital Necker-Enfants Malades (AP-HP), Paris, France
| | - Christine Bodemer
- Department of Dermatology and Pediatric Dermatology, Hôpital Necker Enfants Malades (AP-HP), Paris, France; Institut Imagine, Université Sorbonne Paris cité, Hôpital Necker-Enfants Malades (AP-HP), Paris, France
| | - Karine Briot
- Department of Rheumatology, Hôpital Cochin, Paris Descartes University, Paris, France
| | - Laurent Frenzel
- Department of Hematology, Hôpital Necker EnfantsMalades (AP-HP), Paris, France; Institut Imagine, Université Sorbonne Paris cité, Hôpital Necker-Enfants Malades (AP-HP), Paris, France
| | - Olivier Lortholary
- Department of Infectious Diseases and Tropical Medicine, Hôpital Necker Enfants Malades (AP-HP), Paris, France; Institut Imagine, Université Sorbonne Paris cité, Hôpital Necker-Enfants Malades (AP-HP), Paris, France
| | - Marie-Olivia Chandesris
- Department of Hematology, Hôpital Necker EnfantsMalades (AP-HP), Paris, France; Institut Imagine, Université Sorbonne Paris cité, Hôpital Necker-Enfants Malades (AP-HP), Paris, France
| | - Olivier Hermine
- Department of Hematology, Hôpital Necker EnfantsMalades (AP-HP), Paris, France; Institut Imagine, Université Sorbonne Paris cité, Hôpital Necker-Enfants Malades (AP-HP), Paris, France
| | | |
Collapse
|
42
|
Suurmond J, Rivellese F, Dorjée AL, Bakker AM, Rombouts YJPC, Rispens T, Wolbink G, Zaldumbide A, Hoeben RC, Huizinga TWJ, Toes REM. Toll-like receptor triggering augments activation of human mast cells by anti-citrullinated protein antibodies. Ann Rheum Dis 2014; 74:1915-23. [PMID: 24818634 DOI: 10.1136/annrheumdis-2014-205562] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/17/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Mast cells may play a role in rheumatoid arthritis (RA), but activation of human mast cells in autoimmune settings has been little studied. Toll-like receptors (TLR) and Fcγ receptors (FcγR) are important receptors for cellular activation in the joint, but expression and stimulation of these receptors in human mast cells or the functional interplay between these pathways is poorly understood. Here, we analysed triggering of human mast cells via these receptors in the context of anti-citrullinated protein antibody-positive (ACPA+) RA. METHODS RNA and protein expression of TLRs and FcγR was quantified using PCR and flow cytometry, respectively. Mast cells were stimulated with TLR ligands (including HSP70) combined with IgG immune complexes and IgG-ACPA. RESULTS Human mast cells expressed TLRs and produced cytokines in response to TLR ligands. Both cultured and synovial mast cells expressed FcγRIIA, and triggering of this receptor by IgG immune complexes synergised with activation by TLR ligands, leading to two- to fivefold increased cytokine levels. Mast cells produced cytokines in response to ACPA immune complexes in a citrulline-specific manner, which synergised in the presence of HSP70. CONCLUSIONS Our data show that synovial mast cells express FcγRIIA and that mast cells can be activated by IgG-ACPA and TLR ligands. Importantly, combined stimulation via TLRs and immune complexes leads to synergy in cytokine production. These findings suggest mast cells are important targets for TLR ligands and immune complexes, and that combined activation of mast cells via these pathways greatly enhances inflammation in synovial tissue of RA patients.
Collapse
Affiliation(s)
- J Suurmond
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - F Rivellese
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - A L Dorjée
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - A M Bakker
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Y J P C Rombouts
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - T Rispens
- Sanquin Research and Landsteiner Laboratorium, Academic Medical Center, Amsterdam, The Netherlands
| | - G Wolbink
- Sanquin Research and Landsteiner Laboratorium, Academic Medical Center, Amsterdam, The Netherlands
| | - A Zaldumbide
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - R C Hoeben
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - T W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - R E M Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
43
|
Mast cells as a potential prognostic marker in prostate cancer. DISEASE MARKERS 2013; 35:711-20. [PMID: 24324287 PMCID: PMC3844173 DOI: 10.1155/2013/478303] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 10/07/2013] [Indexed: 12/21/2022]
Abstract
Despite years of intensive investigation that has been made in understanding prostate cancer, it remains one of the major men's health issues and the leading cause of death worldwide. It is now ascertained that prostate cancer emerges from multiple spontaneous and/or inherited alterations that induce changes in expression patterns of genes and proteins that function in complex networks controlling critical cellular events. It is now accepted that several innate and adaptive immune cells, including T- and B-lymphocytes, macrophages, natural killer cells, dendritic cells, neutrophils, eosinophils, and mast cells (MCs), infiltrate the prostate cancer. All of these cells are irregularly scattered within the tumor and loaded with an assorted array of cytokines, chemokines, and inflammatory and cytotoxic mediators. This complex framework reflects the diversity in tumor biology and tumor-host interactions. MCs are well-established effector cells in Immunoglobulin-E (Ig-E) associated immune responses and potent effector cells of the innate immune system; however, their clinical significance in prostate cancer is still debated. Here, these controversies are summarized, focusing on the implications of these findings in understanding the roles of MCs in primary prostate cancer.
Collapse
|
44
|
Kashiwakura JI, Yanagisawa M, Lee H, Okamura Y, Sasaki-Sakamoto T, Saito S, Tokuhashi Y, Ra C, Okayama Y. Interleukin-33 synergistically enhances immune complex-induced tumor necrosis factor alpha and interleukin-8 production in cultured human synovium-derived mast cells. Int Arch Allergy Immunol 2013; 161 Suppl 2:32-6. [PMID: 23711851 DOI: 10.1159/000350424] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Substantial evidence suggests that human synovial mast cells (MCs) are involved in the pathogenesis of rheumatoid arthritis (RA). Interleukin (IL)-33 is believed to play an important role in the pathogenesis of RA. We recently reported that FcγRI is responsible for producing abundant tumor necrosis factor alpha (TNF-α) from cultured synovium-derived MCs (SyMCs) in response to aggregated immunoglobulin G (IgG). However, whether or not IL-33 affects immune complex (IC)-induced synovial MC activation remains unknown. This study sought to evaluate the effect of IL-33 on IC-induced synovial MC activation. METHODS Cultured SyMCs were generated by culturing synovial cells with stem cell factor. ST2 expression was analyzed using FACS and immunohistochemical techniques. Mediators released from the MCs were measured using EIAs or ELISAs. RESULTS SyMCs obtained from patients with RA or osteoarthritis (OA) expressed ST2 on their surfaces. We confirmed the expression of ST2 in MCs using immunofluorescence staining in joint tissue obtained from RA patients. IC-triggered histamine release was not enhanced by IL-33. However, IL-33 synergistically enhanced IC-induced IL-8 and TNF-α production in SyMCs. CONCLUSIONS ICs and IL-33 may exacerbate inflammation associated with RA by abundantly producing TNF-α and IL-8 from SyMCs.
Collapse
Affiliation(s)
- Jun-ichi Kashiwakura
- Department of Molecular Cell Immunology and Allergology, Nihon University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|