1
|
Luo S, Xu H, Gong X, Shen J, Chen X, Wu Z. The complement C3a‑C3aR and C5a‑C5aR pathways promote viability and inflammation of human retinal pigment epithelium cells by targeting NF‑κB signaling. Exp Ther Med 2022; 24:493. [PMID: 35837068 PMCID: PMC9257899 DOI: 10.3892/etm.2022.11420] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/08/2022] [Indexed: 11/05/2022] Open
Abstract
Retinal detachment (RD) and its special form of rhegmatogenous RD associated with choroidal detachment (RRDCD) feature similar pathological alterations, including enhanced retinal cell inflammation. Although the importance of the complement components C3a and C5a and their corresponding receptors in retinal maintenance has been demonstrated, the relevance of these molecules to the pathogenesis of RD or RRDCD remains to be investigated. The contents of C3a, C5a and inflammatory factors, such as TNF-α, IL-1β, IL-6 and prostaglandin (PG)E2, in related clinical samples were examined by ELISA. Subsequently, human retinal pigment epithelial (HRPE) cells were subjected to challenge with the C3a and C5a recombinant proteins with or without C3a and C5a antagonists and NF-κB inhibitor, and the cell viability and inflammatory cytokines were then determined by a Cell Counting Kit-8 assay and ELISA, respectively. In addition, reverse transcription-quantitative PCR and western blot analyses were utilized to examine the mRNA or/and protein levels of C3a and its receptor C3aR, as well as C5a and its receptor C5aR, and NF-κB. In addition, the correlation of C3a and C5a with the aforementioned inflammatory factors was analyzed. The inflammatory factor levels of C3a and C5a were considerably elevated in patients with RRDCD compared to those in the controls. Consistently, C3a and C5a treatment led to increased cell viability and aggravated inflammation in HRPE cells. Accordingly, C3a and C5a induced upregulation of their corresponding receptors C3aR and C5aR, which was in turn observed to be linked to the activation of the NF-κB signaling pathway. Furthermore, there was a positive correlation of the complements C3a and C5a with individual TNF-α, IL-1β, IL-6 and PGE2. Taken together, the C3a-C3aR and C5a-C5aR pathways were indicated to promote cell viability and inflammation of HRPE cells by targeting the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Shasha Luo
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, Jiangsu 214002, P.R. China
| | - Huiyan Xu
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, Jiangsu 214002, P.R. China
| | - Xuechun Gong
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, Jiangsu 214002, P.R. China
| | - Jinyan Shen
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, Jiangsu 214002, P.R. China
| | - Xuan Chen
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, Jiangsu 214002, P.R. China
| | - Zhifeng Wu
- Department of Ophthalmology, Nanjing Medical University Affiliated Wuxi Second Hospital, Wuxi, Jiangsu 214002, P.R. China
| |
Collapse
|
2
|
Westacott LJ, Humby T, Haan N, Brain SA, Bush EL, Toneva M, Baloc AI, Moon AL, Reddaway J, Owen MJ, Hall J, Hughes TR, Morgan BP, Gray WP, Wilkinson LS. Complement C3 and C3aR mediate different aspects of emotional behaviours; relevance to risk for psychiatric disorder. Brain Behav Immun 2022; 99:70-82. [PMID: 34543680 DOI: 10.1016/j.bbi.2021.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 12/18/2022] Open
Abstract
Complement is a key component of the immune system with roles in inflammation and host-defence. Here we reveal novel functions of complement pathways impacting on emotional reactivity of potential relevance to the emerging links between complement and risk for psychiatric disorder. We used mouse models to assess the effects of manipulating components of the complement system on emotionality. Mice lacking the complement C3a Receptor (C3aR-/-) demonstrated a selective increase in unconditioned (innate) anxiety whilst mice deficient in the central complement component C3 (C3-/-) showed a selective increase in conditioned (learned) fear. The dissociable behavioural phenotypes were linked to different signalling mechanisms. Effects on innate anxiety were independent of C3a, the canonical ligand for C3aR, consistent with the existence of an alternative ligand mediating innate anxiety, whereas effects on learned fear were due to loss of iC3b/CR3 signalling. Our findings show that specific elements of the complement system and associated signalling pathways contribute differentially to heightened states of anxiety and fear commonly seen in psychopathology.
Collapse
Affiliation(s)
- Laura J Westacott
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Trevor Humby
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Niels Haan
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK
| | - Sophie A Brain
- Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Emma-Louise Bush
- Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Margarita Toneva
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Andreea-Ingrid Baloc
- Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Anna L Moon
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK
| | - Jack Reddaway
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Michael J Owen
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - Timothy R Hughes
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XW, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK
| | - B Paul Morgan
- Complement Biology Group, Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff CF14 4XW, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK; UK Dementia Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - William P Gray
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK; Brain Repair and Intracranial Therapeutics (BRAIN) Unit, School of Medicine, Cardiff University, CF24 4HQ, UK
| | - Lawrence S Wilkinson
- Neuroscience and Mental Health Research Institute, MRC Centre for Neuropsychiatric Genetic and Genomics, School of Medicine, Hadyn Ellis Building, Cardiff University, Cardiff CF24 4HQ, UK; Behavioural Genetics Group, Schools of Psychology and Medicine, Cardiff University, Cardiff CF10 3AT, UK; Hodge Centre for Neuropsychiatric Immunology, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK.
| |
Collapse
|
3
|
Ligands and Signaling of Mas-Related G Protein-Coupled Receptor-X2 in Mast Cell Activation. Rev Physiol Biochem Pharmacol 2021; 179:139-188. [PMID: 33479839 DOI: 10.1007/112_2020_53] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mas-related G protein-coupled receptor-X2 (MRGPRX2) is known as a novel receptor to activate mast cells (MCs). MRGPRX2 plays a dual role in promoting MC-dependent host defense and immunomodulation and contributing to the pathogenesis of pseudo-allergic drug reactions, pain, itching, and inflammatory diseases. In this article, we discuss the possible signaling pathways of MCs activation mediated by MRGPRX2 and summarize and classify agonists and inhibitors of MRGPRX2 in MCs activation. MRGPRX2 is a low-affinity and low-selectivity receptor, which allows it to interact with a diverse group of ligands. Diverse MRGPRX2 ligands utilize conserved residues in its transmembrane (TM) domains and carboxyl-terminus Ser/Thr residues to undergo ligand binding and G protein coupling. The coupling likely initiates phosphorylation cascades, induces Ca2+ mobilization, and causes degranulation and generation of cytokines and chemokines via MAPK and NF-κB pathways, resulting in MCs activation. Agonists of MRGPRX2 on MCs are divided into peptides (including antimicrobial peptides, neuropeptides, MC degranulating peptides, peptide hormones) and nonpeptides (including FDA-approved drugs). Inhibitors of MRGPRX2 include non-selective GPCR inhibitors, herbal extracts, small-molecule MRGPRX2 antagonists, and DNA aptamer drugs. Screening and classifying MRGPRX2 ligands and summarizing their signaling pathways would improve our understanding of MRGPRX2-mediated physiological and pathological effects on MCs.
Collapse
|
4
|
West PW, Bahri R, Garcia-Rodriguez KM, Sweetland G, Wileman G, Shah R, Montero A, Rapley L, Bulfone-Paus S. Interleukin-33 Amplifies Human Mast Cell Activities Induced by Complement Anaphylatoxins. Front Immunol 2021; 11:615236. [PMID: 33597949 PMCID: PMC7882629 DOI: 10.3389/fimmu.2020.615236] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022] Open
Abstract
Both, aberrant mast cell responses and complement activation contribute to allergic diseases. Since mast cells are highly responsive to C3a and C5a, while Interleukin-33 (IL-33) is a potent mast cell activator, we hypothesized that IL-33 critically regulates mast cell responses to complement anaphylatoxins. We sought to understand whether C3a and C5a differentially activate primary human mast cells, and probe whether IL-33 regulates C3a/C5a-induced mast cell activities. Primary human mast cells were generated from peripheral blood precursors or isolated from healthy human lung tissue, and mast cell complement receptor expression, degranulation, mediator release, phosphorylation patterns, and calcium flux were assessed. Human mast cells of distinct origin express constitutively higher levels of C3aR1 than C5aR1, and both receptors are downregulated by anaphylatoxins. While C3a is a potent mast cell degranulation inducer, C5a is a weaker secretagogue with more delayed effects. Importantly, IL-33 potently enhances the human mast cell reactivity to C3a and C5a (degranulation, cytokine and chemokine release), independent of changes in C3a or C5a receptor expression or the level of Ca2+ influx. Instead, this reflects differential dynamics of intracellular signaling such as ERK1/2 phosphorylation. Since primary human mast cells respond differentially to anaphylatoxin stimulation, and that IL-33 is a key regulator of mast cell responses to complement anaphylatoxins, this is likely to aggravate Th2 immune responses. This newly identified cross-regulation may be important for controlling exacerbated complement- and mast cell-dependent Th2 responses and thus provides an additional rationale for targeting anti-IL33 therapeutically in allergic diseases.
Collapse
Affiliation(s)
- Peter W. West
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Rajia Bahri
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Karen M. Garcia-Rodriguez
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Georgia Sweetland
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Georgia Wileman
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Rajesh Shah
- Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, United Kingdom
| | - Angeles Montero
- Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, United Kingdom
| | - Laura Rapley
- Adaptive Immunity, GlaxoSmithKline, Stevenage, United Kingdom
| | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom,*Correspondence: Silvia Bulfone-Paus,
| |
Collapse
|
5
|
Han R, Hu S, Qin W, Shi J, Hou Q, Wang X, Xu X, Zhang M, Zeng C, Liu Z, Bao H. C3a and suPAR drive versican V1 expression in tubular cells of focal segmental glomerulosclerosis. JCI Insight 2019; 4:122912. [PMID: 30944246 DOI: 10.1172/jci.insight.122912] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 02/14/2019] [Indexed: 12/13/2022] Open
Abstract
Chronic tubulointerstitial injury impacts the prognosis of focal segmental glomerulosclerosis (FSGS). We found that the level of versican V1 was increased in tubular cells of FSGS patients. Tubular cell-derived versican V1 induced proliferation and collagen synthesis by activating the CD44/Smad3 pathway in fibroblasts. Both urine C3a and suPAR were increased and bound to the tubular cells in FSGS patients. C3a promoted the transcription of versican by activating the AKT/β-catenin pathway. C3aR knockout decreased the expression of versican in Adriamycin-treated (ADR-treated) mice. On the other hand, suPAR bound to integrin β6 and activated Rac1, which bound to SRp40 at the 5' end of exon 7 in versican pre-mRNA. This binding inhibited the 3'-end splicing of intron 6 and the base-pair interactions between intron 6 and intron 8, leading to the formation of versican V1. Cotreatment with ADR and suPAR specifically increased the level of versican V1 in tubulointerstitial tissues and caused more obvious interstitial fibrosis in mice than treatment with only ADR. Altogether, our results show that C3a and suPAR drive versican V1 expression in tubular cells by promoting transcription and splicing, respectively, and the increases in tubular cell-derived versican V1 induce interstitial fibrosis by activating fibroblasts in FSGS.
Collapse
Affiliation(s)
- Runhong Han
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Southeast University School of Medicine, Nanjing, China
| | - Shuai Hu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Weisong Qin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jinsong Shi
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Qin Hou
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xia Wang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Minchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Caihong Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Southeast University School of Medicine, Nanjing, China
| | - Hao Bao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
6
|
Quell KM, Karsten CM, Kordowski A, Almeida LN, Briukhovetska D, Wiese AV, Sun J, Ender F, Antoniou K, Schröder T, Schmudde I, Berger JL, König P, Vollbrandt T, Laumonnier Y, Köhl J. Monitoring C3aR Expression Using a Floxed tdTomato-C3aR Reporter Knock-in Mouse. THE JOURNAL OF IMMUNOLOGY 2017. [PMID: 28626064 DOI: 10.4049/jimmunol.1700318] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
C3a exerts multiple biologic functions through activation of its cognate C3a receptor. C3-/- and C3aR-/- mice have been instrumental in defining important roles of the C3a/C3aR axis in the regulation of acute and chronic inflammatory diseases, including ischemia/reperfusion injury, allergic asthma, autoimmune nephritis, and rheumatoid arthritis. Surprisingly little is known about C3aR expression and function in immune and stromal cells. To close this gap, we generated a floxed tandem-dye Tomato (tdTomato)-C3aR reporter knock-in mouse, which we used to monitor C3aR expression in cells residing in the lung, airways, lamina propria (LP) of the small intestine, brain, visceral adipose tissue, bone marrow (BM), spleen, and the circulation. We found a strong expression of tdTomato-C3aR in the brain, lung, LP, and visceral adipose tissue, whereas it was minor in the spleen, blood, BM, and the airways. Most macrophage and eosinophil populations were tdTomato-C3aR+ Interestingly, most tissue eosinophils and some macrophage populations expressed C3aR intracellularly. BM-derived dendritic cells (DCs), lung-resident cluster of differentiation (CD) 11b+ conventional DCs (cDCs) and monocyte-derived DCs, LP CD103+, and CD11b+ cDCs but not pulmonary CD103+ cDCs and splenic DCs were tdTomato-C3aR+ Surprisingly, neither BM, blood, lung neutrophils, nor mast cells expressed C3aR. Similarly, all lymphoid-derived cells were tdTomato-C3aR-, except some LP-derived type 3 innate lymphoid cells. Pulmonary and LP-derived epithelial cells expressed at best minor levels of C3aR. In summary, we provide novel insights into the expression pattern of C3aR in mice. The floxed C3aR knock-in mouse will help to reliably track and conditionally delete C3aR expression in experimental models of inflammation.
Collapse
Affiliation(s)
- Katharina M Quell
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Anna Kordowski
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | | | - Daria Briukhovetska
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Anna V Wiese
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Jing Sun
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Konstantina Antoniou
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Torsten Schröder
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Inken Schmudde
- Institute of Anatomy, University of Lübeck, Lübeck 23562, Germany
| | - Johann L Berger
- Institute of Anatomy, University of Lübeck, Lübeck 23562, Germany
| | - Peter König
- Institute of Anatomy, University of Lübeck, Lübeck 23562, Germany
| | - Tillman Vollbrandt
- Cell Analysis Core Facility, University of Lübeck, Lübeck 23562, Germany; and
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany;
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany; .,Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| |
Collapse
|
7
|
Novel insights into the expression pattern of anaphylatoxin receptors in mice and men. Mol Immunol 2017; 89:44-58. [PMID: 28600003 DOI: 10.1016/j.molimm.2017.05.019] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 02/06/2023]
Abstract
The anaphylatoxins (AT) C3a and C5a play important roles as mediators of inflammation. Further, they regulate and control multiple innate and adaptive immune responses through binding and activation of their cognate G protein-coupled receptors, i.e. C3a receptor (C3aR), C5a receptor 1 (C5aR1) and C5a receptor 2 (C5aR2), although the latter lacks important sequence motifs for G protein-coupling. Based on their pleiotropic functions, they contribute not only to tissue homeostasis but drive, perpetuate and resolve immune responses in many inflammatory diseases including infections, malignancies, autoimmune as well as allergic diseases. During the past few years, transcriptome expression data provided detailed insights into AT receptor tissue mRNA expression. In contrast, our understanding of cellular AT receptor expression in human and mouse tissues under steady and inflammatory conditions is still sketchy. Ligand binding studies, flow cytometric and immunohistochemical analyses convincingly demonstrated tissue-specific C5aR1 expression in various cells of myeloid origin. However, a detailed map for C3aR or C5aR2 expression in human or mouse tissue cells is still lacking. Also, reports about AT expression in lymphoid cells is still controversial. To understand the multiple roles of the ATs in the innate and adaptive immune networks, a detailed understanding of their receptor expression in health and disease is required. Recent findings obtained with novel GFP or tdTomato AT-receptor knock-in mice provide detailed insights into their expression pattern in tissue immune and stroma cells. Here, we will provide an update about our current knowledge of AT receptor expression pattern in humans and mice.
Collapse
|
8
|
Kikuchi-Ueda T, Kamoshida G, Ubagai T, Nakano R, Nakano A, Akuta T, Hikosaka K, Tansho-Nagakawa S, Kikuchi H, Ono Y. The TNF-α of mast cells induces pro-inflammatory responses during infection with Acinetobacter baumannii. Immunobiology 2017; 222:1025-1034. [PMID: 28595750 DOI: 10.1016/j.imbio.2017.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/18/2017] [Accepted: 05/23/2017] [Indexed: 01/12/2023]
Abstract
Mast cells serve important roles as sentinels against bacterial infection by secreting mediators stored in granules. Much of their effectiveness depends upon recruiting and/or modulating other immune cells. The location of mast cells implies that they recognize pathogens invading tissues or mucosal tissues. Acinetobacter baumannii is a gram-negative bacterium that is considered an emerging nosocomial pathogen and causes a wide range of infections associated with high morbidity and mortality. To date, the interaction of A. baumannii with mast cells remains unclear. In this study, we demonstrated an interaction between human LAD2 mast cells and A. baumannii in vitro. When LAD2 cells were co-cultured with live A. baumannii or Pseudomonas aeruginosa PAO1 in vitro for 4h, TNF-α and IL-8 were produced in the culture supernatant. These inflammatory cytokines were not detected in the supernatant after the cells were treated with live bacteria without serum. Gene expression analysis showed that TNF-α and IL-8 mRNA expression increased in A. baumannii- and P. aeruginosa-infected LAD2 cells. Scanning electron microscopy showed that A. baumannii was tightly attached to the surface of LAD2 cells and suggested that A. baumannii may bind to FcγRII (CD32) on LAD2 cells. TNF-α in the culture supernatant from A. baumannii-infected LAD2 cells, showed that PMN activation and migration increased in Boyden chamber assays. These results suggest that mast cells recognize and initiate immune responses toward A. baumannii by releasing the preformed mediator TNF-α to activate effector neutrophils.
Collapse
Affiliation(s)
- Takane Kikuchi-Ueda
- Department of Microbiology and Immunology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Go Kamoshida
- Department of Microbiology and Immunology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Tsuneyuki Ubagai
- Department of Microbiology and Immunology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Ryuichi Nakano
- Department of Microbiology and Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara-shi, Nara 634-8521, Japan.
| | - Akiyo Nakano
- Department of Microbiology and Infectious Diseases, Nara Medical University, 840 Shijo-cho, Kashihara-shi, Nara 634-8521, Japan.
| | - Teruo Akuta
- Department of Microbiology and Immunology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Kenji Hikosaka
- Department of Infection and Host Defense, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.
| | - Shigeru Tansho-Nagakawa
- Department of Microbiology and Immunology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Hirotoshi Kikuchi
- Department of Microbiology and Immunology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Yasuo Ono
- Department of Microbiology and Immunology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| |
Collapse
|
9
|
Sheen JH, Strainic MG, Liu J, Zhang W, Yi Z, Medof ME, Heeger PS. TLR-Induced Murine Dendritic Cell (DC) Activation Requires DC-Intrinsic Complement. THE JOURNAL OF IMMUNOLOGY 2017; 199:278-291. [PMID: 28539427 DOI: 10.4049/jimmunol.1700339] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/04/2017] [Indexed: 01/04/2023]
Abstract
Induction of proinflammatory T cell immunity is augmented by innate dendritic cell (DC) maturation commonly initiated by TLR signaling. We demonstrate that ligation of TLR3, TLR4, and TLR9 induces murine DC production of complement components and local production of the anaphylatoxin C5a. In vitro, ex vivo, and in vivo analyses show that TLR-induced DC maturation, as assessed by surface phenotype, expression profiling by gene array, and functional ability to stimulate T cell responses, requires autocrine C3a receptor and C5a receptor (C3ar1/C5ar1) signaling. Studies using bone marrow chimeric animals and Foxp3-GFP/ERT2-Cre/dTomato fate-mapping mice show that TLR-initiated DC autocrine C3ar1/C5ar1 signaling causes expansion of effector T cells and instability of regulatory T cells and contributes to T cell-dependent transplant rejection. Together, our data position immune cell-derived complement production and autocrine/paracrine C3ar1/C5ar1 signaling as crucial intermediary processes that link TLR stimulation to DC maturation and the subsequent development of effector T cell responses.
Collapse
Affiliation(s)
- Joong-Hyuk Sheen
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Institute of Immunology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| | - Michael G Strainic
- Institute of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Jinbo Liu
- Institute of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Weijia Zhang
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Zhengzi Yi
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - M Edward Medof
- Institute of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Peter S Heeger
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029; .,Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029.,Precision Institute of Immunology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| |
Collapse
|
10
|
Abstract
Mast cells (MCs) play a central role in tissue homoeostasis, sensing the local environment through numerous innate cell surface receptors. This enables them to respond rapidly to perceived tissue insults with a view to initiating a co-ordinated programme of inflammation and repair. However, when the tissue insult is chronic, the ongoing release of multiple pro-inflammatory mediators, proteases, cytokines and chemokines leads to tissue damage and remodelling. In asthma, there is strong evidence of ongoing MC activation, and their mediators and cell-cell signals are capable of regulating many facets of asthma pathophysiology. This article reviews the evidence behind this.
Collapse
Affiliation(s)
- P Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| | - G Arthur
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| |
Collapse
|
11
|
Subramanian H, Gupta K, Ali H. Roles of Mas-related G protein-coupled receptor X2 on mast cell-mediated host defense, pseudoallergic drug reactions, and chronic inflammatory diseases. J Allergy Clin Immunol 2016; 138:700-710. [PMID: 27448446 DOI: 10.1016/j.jaci.2016.04.051] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/21/2016] [Accepted: 04/06/2016] [Indexed: 12/11/2022]
Abstract
Mast cells (MCs), which are granulated tissue-resident cells of hematopoietic lineage, contribute to vascular homeostasis, innate/adaptive immunity, and wound healing. However, MCs are best known for their roles in allergic and inflammatory diseases, such as anaphylaxis, food allergy, rhinitis, itch, urticaria, atopic dermatitis, and asthma. In addition to the high-affinity IgE receptor (FcεRI), MCs express numerous G protein-coupled receptors (GPCRs), which are the largest group of membrane receptor proteins and the most common targets of drug therapy. Antimicrobial host defense peptides, neuropeptides, major basic protein, eosinophil peroxidase, and many US Food and Drug Administration-approved peptidergic drugs activate human MCs through a novel GPCR known as Mas-related G protein-coupled receptor X2 (MRGPRX2; formerly known as MrgX2). Unique features of MRGPRX2 that distinguish it from other GPCRs include their presence both on the plasma membrane and intracellular sites and their selective expression in MCs. In this article we review the possible roles of MRGPRX2 on host defense, drug-induced anaphylactoid reactions, neurogenic inflammation, pain, itch, and chronic inflammatory diseases, such as urticaria and asthma. We propose that host defense peptides that kill microbes directly and activate MCs through MRGPRX2 could serve as novel GPCR targets to modulate host defense against microbial infection. Furthermore, mAbs or small-molecule inhibitors of MRGPRX2 could be developed for the treatment of MC-dependent allergic and inflammatory disorders.
Collapse
Affiliation(s)
- Hariharan Subramanian
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pa
| | - Kshitij Gupta
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pa
| | - Hydar Ali
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pa.
| |
Collapse
|
12
|
Lian H, Yang L, Cole A, Sun L, Chiang ACA, Fowler SW, Shim DJ, Rodriguez-Rivera J, Taglialatela G, Jankowsky JL, Lu HC, Zheng H. NFκB-activated astroglial release of complement C3 compromises neuronal morphology and function associated with Alzheimer's disease. Neuron 2014; 85:101-115. [PMID: 25533482 DOI: 10.1016/j.neuron.2014.11.018] [Citation(s) in RCA: 446] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2014] [Indexed: 11/26/2022]
Abstract
Abnormal NFκB activation has been implicated in Alzheimer's disease (AD). However, the signaling pathways governing NFκB regulation and function in the brain are poorly understood. We identify complement protein C3 as an astroglial target of NFκB and show that C3 release acts through neuronal C3aR to disrupt dendritic morphology and network function. Exposure to Aβ activates astroglial NFκB and C3 release, consistent with the high levels of C3 expression in brain tissue from AD patients and APP transgenic mice, where C3aR antagonist treatment rescues cognitive impairment. Therefore, dysregulation of neuron-glia interaction through NFκB/C3/C3aR signaling may contribute to synaptic dysfunction in AD, and C3aR antagonists may be therapeutically beneficial.
Collapse
Affiliation(s)
- Hong Lian
- Huffington Center on Aging, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Houston, TX 77030, USA
| | - Li Yang
- Huffington Center on Aging, Houston, TX 77030, USA
| | - Allysa Cole
- Huffington Center on Aging, Houston, TX 77030, USA
| | - Lu Sun
- Huffington Center on Aging, Houston, TX 77030, USA
| | - Angie C-A Chiang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephanie W Fowler
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - David J Shim
- Huffington Center on Aging, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Giulio Taglialatela
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Joanna L Jankowsky
- Huffington Center on Aging, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hui-Chen Lu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine and the Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Hui Zheng
- Huffington Center on Aging, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Cudaback E, Yang Y, Montine TJ, Keene CD. APOE genotype-dependent modulation of astrocyte chemokine CCL3 production. Glia 2014; 63:51-65. [PMID: 25092803 DOI: 10.1002/glia.22732] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 07/14/2014] [Indexed: 12/19/2022]
Abstract
Apolipoprotein E (apoE) is well known as a regulator of cholesterol homeostasis, and is increasingly recognized to play a prominent role in the modulation of innate immune response, including cell-to-cell communication and migration. Alzheimer's disease (AD) is a slowly progressive neurodegenerative disorder characterized by neuroinflammation that appears to be an important component of the pathophysiology of the disease. Astrocytes are the majority cell type in brain, exerting significant influence over a range of central nervous system activities, including microglial-mediated neuroinflammatory responses. As the resident innate immune effector cells of the brain, microglia respond to soluble chemical signals released from tissue during injury and disease by mobilizing to lesion sites, clearing toxic molecules, and releasing chemical signals of their own. While microglial-mediated neuroinflammation in the AD brain remains an area of intense investigation, the mechanisms underlying reinforcement and regulation of these aberrant microglial responses by astrocytes are largely unstudied. Moreover, although inheritance of APOE ɛ4 represents the greatest genetic risk factor for sporadic AD, the mechanism by which apoE isoforms differentially influence AD pathophysiology is unknown. Here we show that APOE ɛ4 genotype specifically modulates astrocyte secretion of potent microglial chemotactic agents, including CCL3, thus providing evidence that APOE modulation of central nervous system (CNS) innate immune response is mediated through astrocytes.
Collapse
Affiliation(s)
- Eiron Cudaback
- Department of Pathology, University of Washington, Seattle, Washington
| | | | | | | |
Collapse
|
14
|
Stefos GC, Soppa U, Dierssen M, Becker W. NGF upregulates the plasminogen activation inhibitor-1 in neurons via the calcineurin/NFAT pathway and the Down syndrome-related proteins DYRK1A and RCAN1 attenuate this effect. PLoS One 2013; 8:e67470. [PMID: 23825664 PMCID: PMC3692457 DOI: 10.1371/journal.pone.0067470] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 05/18/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Plasminogen activator inhibitor 1 (PAI-1) is a key regulator of the plasminogen activation system. Although several lines of evidence support a significant role of PAI-1 in the brain, the regulation of its expression in neurons is poorly understood. In the present study we tested the hypothesis that NGF induces the upregulation of PAI-1 via the calcineurin/nuclear factor of activated T cells (NFAT) pathway and analysed whether the overexpression of the Down syndrome-related proteins DYRK1A and RCAN1 modulated the effect of NGF on PAI-1 expression. RESULTS NGF upregulated PAI-1 mRNA levels in primary mouse hippocampal neurons cultured for 3 days in vitro and in the rat pheochromocytoma cell line PC12. Reporter gene assays revealed that NGF activated the calcineurin/NFAT pathway in PC12 cells. Induction of PAI-1 by NGF was sensitive to the calcineurin inhibitor FK506 and the specific inhibition of NFAT activation by the cell permeable VIVIT peptide. Activation of calcineurin/NFAT signalling through other stimuli resulted in a much weaker induction of PAI-1 expression, suggesting that other NGF-induced pathways are involved in PAI-1 upregulation. Overexpression of either DYRK1A or RCAN1 negatively regulated NFAT-dependent transcriptional activity and reduced the upregulation of PAI-1 levels by NGF. CONCLUSION The present results show that the calcineurin/NFAT pathway mediates the upregulation of PAI-1 by NGF. The negative effect of DYRK1A and RCAN1 overexpression on NGF signal transduction in neural cells may contribute to the altered neurodevelopment and brain function in Down syndrome.
Collapse
Affiliation(s)
- Georgios C Stefos
- Institute of Pharmacology and Toxicology, Medical Faculty of the RWTH Aachen University, Aachen, Germany.
| | | | | | | |
Collapse
|
15
|
Yu M, Zou W, Peachey NS, McIntyre TM, Liu J. A novel role of complement in retinal degeneration. Invest Ophthalmol Vis Sci 2012; 53:7684-92. [PMID: 23074214 DOI: 10.1167/iovs.12-10069] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The association of single nucleotide polymorphisms of components of the complement alternative pathway with the risk of age-related macular degeneration (AMD) indicates that complement signaling plays an important role in retinal physiology. How genetic variation leads to retinal degeneration is unknown. It has been assumed that complement activation augments immune responses, which in turn initiate AMD pathogenesis. To better understand the relationship between complement and the outer retina, we examined mice lacking the main complement component C3 and the receptors for complement activation fragments C3a (C3aR) and/or C5a (C5aR). METHODS Complement mutant mice were studied along with wild-type (WT) littermates from 6 weeks to 14 months of age. Strobe flash electroretinography (ERG) was used to examine outer retinal function and a dc-ERG technique was used to measure ERG components generated by the retinal pigment epithelium. Retinas were examined by histology, immunohistochemistry, and biochemistry. RESULTS Mice lacking C3aR and/or C5aR developed early onset and progressive retinal degeneration, accompanied by cleaved caspase-3 upregulation. Genetic deletion of C3aR and/or C5aR led to cell-specific defects that matched the cellular localization of these receptors in the WT retina. Compared to WT, C3aR(-/-) and C3aR(-/-)C5aR(-/-) mice showed increased retinal dysfunction upon light exposure. C3aR(-/-)C5aR(-/-) mice immunized with 4-hydroxynonenal-adducted protein developed severe retinal impairment unrelated to immune response. CONCLUSIONS C3aR- and C5aR-mediated signaling was necessary to maintain normal retinal function and structure. These receptors may be important biomarkers for predicting retinal degeneration including AMD.
Collapse
Affiliation(s)
- Minzhong Yu
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
16
|
Regulation of avoidant behaviors and pain by the anti-inflammatory tyrosine phosphatase SHP-1. ACTA ACUST UNITED AC 2012; 2:235-46. [PMID: 18250891 DOI: 10.1017/s1740925x07000476] [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] [Indexed: 01/05/2023]
Abstract
The protein tyrosine phosphatase SHP-1 is a critical regulator of cytokine signaling and inflammation. Mice homozygous for a null allele at the SHP-1 locus have a phenotype of severe inflammation and are hyper-responsive to the TLR4 ligand LPS. TLR4 stimulation in the CNS has been linked to both neuropathic pain and sickness behaviors. To determine if reduction in SHP-1 expression affects LPS-induced behaviors, responses of heterozygous SHP-1-deficient (me/+) and wild-type (+/+) mice to LPS were measured. Chronic (4-week) treatment with LPS induced avoidant behaviors indicative of fear/anxiety in me/+, but not +/+, mice. These behaviors were correlated with a LPS-induced type 2 cytokine, cytokine receptor, and immune effector arginase profile in the brains of me/+ mice not found in +/+ mice. Me/+ mice also had a constitutively greater level of TLR4 in the CNS than +/+ mice. Additionally, me/+ mice displayed constitutively increased thermal sensitivity compared to +/+ mice, measured by the tail-flick test. Moreover, me/+ glial cultures were more responsive to LPS than +/+ glia. Therefore, the reduced expression of SHP-1 in me/+ imparts haploinsufficiency with respect to the control of CNS TLR4 and pain signaling. Furthermore, type 2 cytokines become prevalent during chronic TLR4 hyperstimulation in the CNS and are associated positively with behaviors that are usually linked to type 1 pro-inflammatory cytokines. These findings question the notion that type 2 immunity is solely anti-inflammatory in the CNS and indicate that type 2 immunity induces/potentiates CNS inflammatory processes.
Collapse
|
17
|
Guo Q, Subramanian H, Gupta K, Ali H. Regulation of C3a receptor signaling in human mast cells by G protein coupled receptor kinases. PLoS One 2011; 6:e22559. [PMID: 21799898 PMCID: PMC3143157 DOI: 10.1371/journal.pone.0022559] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 06/27/2011] [Indexed: 12/03/2022] Open
Abstract
Background The complement component C3a activates human mast cells via its cell surface G protein coupled receptor (GPCR) C3aR. For most GPCRs, agonist-induced receptor phosphorylation leads to receptor desensitization, internalization as well as activation of downstream signaling pathways such as ERK1/2 phosphorylation. Previous studies in transfected COS cells overexpressing G protein coupled receptor kinases (GRKs) demonstrated that GRK2, GRK3, GRK5 and GRK6 participate in agonist-induced C3aR phosphorylation. However, the roles of these GRKs on the regulation of C3aR signaling and mediator release in human mast cells remain unknown. Methodology/Principal Findings We utilized lentivirus short hairpin (sh)RNA to stably knockdown the expression of GRK2, GRK3, GRK5 and GRK6 in human mast cell lines, HMC-1 and LAD2, that endogenously express C3aR. Silencing GRK2 or GRK3 expression caused a more sustained Ca2+ mobilization, attenuated C3aR desensitization, and enhanced degranulation as well as ERK1/2 phosphorylation when compared to shRNA control cells. By contrast, GRK5 or GRK6 knockdown had no effect on C3aR desensitization, but caused a significant decrease in C3a-induced mast cell degranulation. Interestingly, GRK5 or GRK6 knockdown rendered mast cells more responsive to C3a for ERK1/2 phosphorylation. Conclusion/Significance This study demonstrates that GRK2 and GRK3 are involved in C3aR desensitization. Furthermore, it reveals the novel finding that GRK5 and GRK6 promote C3a-induced mast cell degranulation but inhibit ERK1/2 phosphorylation via C3aR desensitization-independent mechanisms. These findings thus reveal a new level of complexity for C3aR regulation by GRKs in human mast cells.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Hariharan Subramanian
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kshitij Gupta
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Hydar Ali
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
18
|
Kashem SW, Subramanian H, Collington SJ, Magotti P, Lambris JD, Ali H. G protein coupled receptor specificity for C3a and compound 48/80-induced degranulation in human mast cells: roles of Mas-related genes MrgX1 and MrgX2. Eur J Pharmacol 2011; 668:299-304. [PMID: 21741965 DOI: 10.1016/j.ejphar.2011.06.027] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 06/08/2011] [Accepted: 06/15/2011] [Indexed: 01/28/2023]
Abstract
Although human mast cells express G protein coupled receptors for the anaphylatoxin C3a, previous studies indicated that C3a causes mast cell degranulation, at least in part, via a C3a receptor-independent mechanism similar to that proposed for polycationic molecules such as compound 48/80. The purpose of the present study was to delineate the receptor specificity of C3a-induced degranulation in human mast cells. We found that C3a, a C3a receptor "superagonist" (E7) and compound 48/80 induced Ca(2+) mobilization and degranulation in a differentiated human mast cell line, LAD2. However, C3a and E7 caused Ca(2+) mobilization in an immature mast cell line, HMC-1 but compound 48/80 did not. We have previously shown that LAD2 cells express MrgX1 and MrgX2 but HMC-1 cells do not. To delineate the receptor specificity for C3a and compound 48/80 further, we generated stable transfectants expressing MrgX1 and MrgX2 in a rodent mast cell line, RBL-2H3 cells. We found that compound 48/80 caused degranulation in RBL-2H3 cells expressing MrgX1 and MrgX2 but C3a did not. By contrast, E7 activated RBL-2H3 cells expressing MrgX2 but not MrgX1. These findings demonstrate that in contrast to previous reports, C3a and compound 48/80 do not use a shared mechanism for mast cell degranulation. It shows that while compound 48/80 utilizes MrgX1 and MrgX2 for mast cell degranulation C3a does not. It further reveals the novel finding that the previously characterized synthetic peptide, C3a receptor "superagonist" E7 activates human mast cells via two mechanisms; one involving the C3a receptor and the other MrgX2.
Collapse
Affiliation(s)
- Sakeen W Kashem
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | | | | | | | | | | |
Collapse
|
19
|
Kim B, Jeong HK, Kim JH, Lee SY, Jou I, Joe EH. Uridine 5'-diphosphate induces chemokine expression in microglia and astrocytes through activation of the P2Y6 receptor. THE JOURNAL OF IMMUNOLOGY 2011; 186:3701-9. [PMID: 21317391 DOI: 10.4049/jimmunol.1000212] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chemokines play critical roles in inflammation by recruiting inflammatory cells to injury sites. In this study, we found that UDP induced expression of chemokines CCL2 (MCP-1) and CCL3 (MIP-1α) in microglia, astrocytes, and slice cultures by activation of P2Y(6). Interestingly, CCL2 was more highly expressed than CCL3. However, CCL2 synthesis kinetics in response to UDP differed in microglia and astrocytes; microglia rapidly produced small amounts of CCL2, whereas astrocytes continuously synthesized large amounts of CCL2, resulting in a high ultimate level of the chemokine. UDP-induced chemokine expression was reduced in the presence of a specific antagonist of P2Y(6) (MRS2578) or small interfering RNA directed against the P2Y(6) gene. Inhibition of phospholipase C and calcium increase, downstream signaling pathways of Gq-coupled P2Y(6), reduced UDP-induced chemokine expression. UDP activated two calcium-activated transcription factors, NFATc1 and c2. Furthermore, inhibitors of calcineurin (a phosphatase activating NFAT) and NFAT reduced UDP-induced chemokine synthesis. We also found, using a transmigration assay, that UDP-treated astrocytes recruited monocytes. These results suggest that UDP induces chemokine expression in microglia and astrocytes of the injured brain by activation of P2Y(6) receptors.
Collapse
Affiliation(s)
- Beomsue Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
Mast cells are multifunctional cells that initiate not only IgE-dependent allergic diseases but also play a fundamental role in innate and adaptive immune responses to microbial infection. They are also thought to play a role in angiogenesis, tissue remodeling, wound healing, and tumor repression or growth. The broad scope of these physiologic and pathologic roles illustrates the flexible nature of mast cells, which is enabled in part by their phenotypic adaptability to different tissue microenvironments and their ability to generate and release a diverse array of bioactive mediators in response to multiple types of cell-surface and cytosolic receptors. There is increasing evidence from studies in cell cultures that release of these mediators can be selectively modulated depending on the types or groups of receptors activated. The intent of this review is to foster interest in the interplay among mast cell receptors to help understand the underlying mechanisms for each of the immunological and non-immunological functions attributed to mast cells. The second intent of this review is to assess the pathophysiologic roles of mast cells and their products in health and disease. Although mast cells have a sufficient repertoire of bioactive mediators to mount effective innate and adaptive defense mechanisms against invading microorganisms, these same mediators can adversely affect surrounding tissues in the host, resulting in autoimmune disease as well as allergic disorders.
Collapse
Affiliation(s)
- Alasdair M Gilfillan
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1881, USA.
| | | |
Collapse
|
21
|
Ali H. Regulation of human mast cell and basophil function by anaphylatoxins C3a and C5a. Immunol Lett 2009; 128:36-45. [PMID: 19895849 DOI: 10.1016/j.imlet.2009.10.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 10/21/2009] [Accepted: 10/23/2009] [Indexed: 12/18/2022]
Abstract
Allergic diseases such as asthma result from inappropriate immunologic responses to common environmental allergens in genetically susceptible individuals. Following allergen exposure, interaction of dendritic cells (DC) with CD4(+) T cells leads to the production of Th2 cytokines, which induce B cells to synthesize IgE molecules (sensitization phase). These IgE molecules bind to their high affinity receptors (FcvarepsilonRI) on the surface of mast cells and basophils and their subsequent cross-linking by allergen results in the release of preformed and newly synthesized mediators, which cause bronchoconstriction, lung inflammation and airway hyperresponsiveness (AHR) in asthma (effector phase). The complement components C3a and C5a levels are increased in the lungs of patients with asthma and are likely generated via the actions of both allergen and mast cell proteases. In vivo studies with rodents have shown that while C3a facilitates allergen sensitization in some models C5a inhibits this response. Despite this difference, both anaphylatoxins promote lung inflammation and AHR in vivo indicating that cells other than DC and T cells likely mediate the functional effects of C3a and C5a in asthma. This review focuses on the contribution of C3a and C5a in the pathogenesis of asthma with a particular emphasis on mast cells and basophils. It discusses the mechanisms by which anaphylatoxins activate mast cells and basophils and the associated signaling pathways via which their receptors are regulated by priming and desensitization.
Collapse
Affiliation(s)
- Hydar Ali
- Department of Pathology, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Philadelphia, PA 19104-6030, USA.
| |
Collapse
|
22
|
Shinjyo N, Ståhlberg A, Dragunow M, Pekny M, Pekna M. Complement-Derived Anaphylatoxin C3a Regulates In Vitro Differentiation and Migration of Neural Progenitor Cells. Stem Cells 2009; 27:2824-32. [DOI: 10.1002/stem.225] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
23
|
Inhibitory effect of glycoprotein isolated from Cudrania tricuspidata bureau on expression of inflammation-related cytokine in bisphenol A-treated HMC-1 cells. Inflammation 2009; 32:211-7. [PMID: 19415476 DOI: 10.1007/s10753-009-9122-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cudrania tricuspidata is one of the most omnipresent traditional herbal drugs for anti-inflammation and anti-tumor. The purpose of the present study was to determine whether the CTB glycoprotein regulates the inflammatory reaction stimulated by bisphenol A (BPA) in human mast cells (HMC-1). Thus, we investigated that CTB glycoprotein inhibits the degranulation of histamine, expression of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), as a mitogen activated protein (MAP) kinase, nuclear transcription factors involving nuclear factor (NF)-kappaB and Activator protein (AP)-1, cyclooxygenase (COX)-2. The results indicated that CTB glycoprotein decreased gene expression of cytokines of IL-4, IFN-gamma, interleukin (IL)-1beta and cyclooxygenase (COX)-2 in BPA-stimulated HMC-1 cells. Hence, we speculate that CTB glycoprotein can use as a potent anti-inflammatory agent for inflammatory allergic diseases.
Collapse
|
24
|
Silver MR, Margulis A, Wood N, Goldman SJ, Kasaian M, Chaudhary D. IL-33 synergizes with IgE-dependent and IgE-independent agents to promote mast cell and basophil activation. Inflamm Res 2009; 59:207-18. [PMID: 19763788 DOI: 10.1007/s00011-009-0088-5] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 08/21/2009] [Accepted: 08/23/2009] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Mast cell and basophil activation contributes to inflammation, bronchoconstriction, and airway hyperresponsiveness in asthma. Because IL-33 expression is inflammation inducible, we investigated IL-33-mediated effects in concert with both IgE-mediated and IgE-independent stimulation. METHODS Because the HMC-1 mast cell line can be activated by GPCR and RTK signaling, we studied the effects of IL-33 on these pathways. The IL-33- and SCF-stimulated HMC-1 cells were co-cultured with human lung fibroblasts and airway smooth muscle cells in a collagen gel contraction assay. IL-33 effects on IgE-mediated activation were studied in primary mast cells and basophils. RESULT IL-33 synergized with adenosine, C5a, SCF, and NGF receptor activation. IL-33-stimulated and SCF-stimulated HMC-1 cells demonstrated enhanced collagen gel contraction when cultured with fibroblasts or smooth muscle cells. IL-33 also synergized with IgE receptor activation of primary human mast cells and basophils. CONCLUSION IL-33 amplifies inflammation in both IgE-independent and IgE-dependent responses.
Collapse
Affiliation(s)
- Matthew R Silver
- Inflammation Research, Wyeth, 200 Cambridge Park Drive, Cambridge, MA, 02140, USA
| | | | | | | | | | | |
Collapse
|
25
|
Wenderfer SE, Wang H, Ke B, Wetsel RA, Braun MC. C3a receptor deficiency accelerates the onset of renal injury in the MRL/lpr mouse. Mol Immunol 2009; 46:1397-404. [PMID: 19167760 DOI: 10.1016/j.molimm.2008.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 12/04/2008] [Accepted: 12/07/2008] [Indexed: 12/23/2022]
Abstract
The development and progression of systemic lupus erythematosus (SLE) is strongly associated with complement activation and deposition. The anaphylatoxin C3a is a product of complement activation with immunomodulatory properties, and the receptor for C3a (C3aR) is not only expressed by granulocytes and antigen presenting cell populations, but it is also strongly up-regulated in lupus prone mice with active nephritis. In order to characterize the role of the C3aR in inflammatory nephritis, we bred C3aR knock out mice onto the MRL/lpr genetic background (C3aR KO MRL). Compared to control MRL/lpr mice, C3aR KO MRL mice had elevated auto-antibody titers and an earlier onset of renal injury. At 8 weeks, renal expression of a wide range of chemokines and chemokine receptors was increased in C3aR KO MRL kidneys compared to controls. Only the expression of MCP-1 was significantly decreased in the C3aR KO MRL mice. The increased chemokine and chemokine receptor expression seen in the C3aR KO MRL mice was associated with a more rapid rise in serum creatinine and the acceleration of renal fibrosis. However, loss of the C3aR had little impact on long-term kidney injury and did not alter survival. These findings suggest that activation of the C3aR plays a protective, not pathologic, role in the early phase of inflammatory nephritis in the MRL/lpr model of SLE.
Collapse
Affiliation(s)
- Scott E Wenderfer
- The Center for Immunology and Autoimmune Diseases, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center-Houston, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
26
|
Kim JY, Kim DY, Ro JY. Granule formation in NGF-cultured mast cells is associated with expressions of pyruvate kinase type M2 and annexin I proteins. Int Arch Allergy Immunol 2008; 146:287-97. [PMID: 18362474 DOI: 10.1159/000121463] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Accepted: 11/27/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Nerve growth factor (NGF) is a potent mediator, which regulates characteristics of mast cells, but its biological function is not well characterized. This study aimed to screen proteins associated with the maturation of human mast cells-1 (HMC-1) or mouse bone marrow-derived mast cells (BMMCs) cultured with NGF, and to examine the functions of proteins involved. METHODS NGF (10 ng/ml) was added to cell culture medium every other day for 10 days for HMC-1 or twice a week for 5 weeks for BMMCs. Granule formation was determined by electron microscopy or May-Grunwald-Giemsa staining, TNF-alpha by ELISA, expressions of various proteins by two-dimensional gel electrophoresis (2-DE), siRNA transfection by Lipofectamine 2000, and the expressions of pyruvate kinase and annexin I by immunoblotting. RESULTS After NGF treatment, granule formation and total amounts of granular mediator, TNF-alpha increased in both mast cells. This TNF-alpha was released by calcium ionophore or by antigen/antibody reaction. Expressions of pyruvate kinase and annexin I obtained by 2-DE were confirmed by immunoblotting and siRNA-transfected HMC-1 cells. Expressions of proteins, granule formation and TNF-alpha content were blocked by both the TrkA inhibitor, K252a, and the ERK inhibitor, PD98059, but not by the PI3 kinase inhibitors, LY294002 and wortmannin. CONCLUSION These data suggest that pyruvate kinase and annexin I expressed by NGF contribute to granule formation containing TNF-alpha as well as other mediators in mast cells, which play a major role in allergic diseases via a TrkA/ERK pathway.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Pharmacology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Korea
| | | | | |
Collapse
|
27
|
Wang J, Hancock MK, Dudek JM, Bi K. Cellular assays for high-throughput screening for modulators of Trk receptor tyrosine kinases. CURRENT CHEMICAL GENOMICS 2008; 1:27-33. [PMID: 20161825 PMCID: PMC2774616 DOI: 10.2174/1875397300801010027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 01/12/2008] [Accepted: 01/21/2008] [Indexed: 11/22/2022]
Abstract
Trk receptor tyrosine kinases are required for signal transduction initiated by neurotrophins leading to cell proliferation, differentiation, survival and death. Alterations in Trk kinase activity have been linked to various diseases. To address the need for cell-based assays for screening and studying the selectivity of Trk kinase modulators, we developed high-throughput cell-based assays for Trk receptor kinases using nuclear factor of activated T-cells (NFAT) beta-lactamase reporter lines stably expressing full length human Trk kinases. These assays were functionally validated with cognate neurotrophin(s), inhibitors and TRK RNAi oligos and demonstrated for their utility in identifying potent and selective modulators of Trk receptor kinases.
Collapse
Affiliation(s)
- Jun Wang
- Invitrogen Corporation, Discovery Sciences, 501 Charmany Drive, Madison, WI 53719, USA
| | | | | | | |
Collapse
|
28
|
Zaidi AK, Ali H. C3a receptors signaling in mast cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 598:126-40. [PMID: 17892209 DOI: 10.1007/978-0-387-71767-8_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Asifa K Zaidi
- University of Pennsylvania School of Dental Medicine, Department of Pathology, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
29
|
Groth RD, Coicou LG, Mermelstein PG, Seybold VS. Neurotrophin activation of NFAT-dependent transcription contributes to the regulation of pro-nociceptive genes. J Neurochem 2007; 102:1162-74. [PMID: 17488277 DOI: 10.1111/j.1471-4159.2007.04632.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) play key roles in the development of inflammation-induced hyperalgesia by triggering the expression of pro-nociceptive genes within primary afferent and spinal neurons. However, the mechanisms by which neurotrophins elicit gene expression remain largely unknown. Recently, neurotrophins have been shown to activate members of the calcineurin (CaN)-regulated, nuclear factor of activated T-cells (NFATc) family of transcription factors within brain. Thus, we hypothesized that NFATc transcription factors couple neurotrophin signaling to gene expression within primary afferent and spinal neurons. In situ hybridization revealed NFATc4 mRNA within the dorsal root ganglion and spinal cord. In cultured dorsal root ganglion cells, NGF triggered NFAT-dependent transcription in a CaN-sensitive manner. Further, increased BDNF expression following NGF treatment relied on CaN, thereby suggesting that NGF regulates BDNF transcription via activation of NFATc4. Within cultured spinal cells, BDNF also activated CaN-dependent, NFAT-regulated gene expression. Interestingly, BDNF stimulation increased the expression of the pro-nociceptive genes cyclooxygenase-2, neurokinin-1 receptor, inositol trisphosphates receptor type 1, and BDNF itself, through both NFAT-dependent and NFAT-independent transcriptional mechanisms. Our results suggest that regulation of pro-nociceptive genes through activation of NFAT-dependent transcription is one mechanism by which NGF and BDNF signaling contributes to the development of persistent pain states.
Collapse
Affiliation(s)
- Rachel D Groth
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|
30
|
Groth RD, Coicou LG, Mermelstein PG, Seybold VS. Neurotrophin activation of NFAT-dependent transcription contributes to the regulation of pro-nociceptive genes. J Neurochem 2007. [DOI: 10.1111/j.1471-4159.2007.4632.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
31
|
Abstract
The complement system is an important component of the innate immune system and a modulator of adaptive immunity. The entire complement system is focused on C3 and C5. Thus, there are proteins that activate C3 and C5, those that regulate this activation, and those that transduce the effects of C3 and C5 activation products; each can affect the kidney in renal injury. The normal kidney has the inherent capacity to protect itself from complement activation through cellular expression of decay-accelerating factor, membrane cofactor protein (in human beings), and Crry (in rodents). In addition, plasma factor H protects vascular spaces in the kidney. Although the main function of these proteins is to limit complement activation, there is now considerable evidence that they can transduce signals on engagement in immune cells. The G-protein-coupled 7-span transmembrane receptors for C3a and C5a, and the integral membrane complement receptors (CR) for C3b, iC3b, and C3dg, are expressed outside the kidney, particularly in cells of hematopoietic and immune lineage. These are important in renal injury through their infiltration of the kidney and/or by affecting kidney-directed immune responses. There is mounting evidence that intrinsic glomerular and tubular cell C3aR and C5aR expression and activation also can affect renal injury. CR1 on podocytes and the beta2 integrins CR3 and CR4 in kidney dendritic cells have functions that remain poorly defined. Cells of the kidney also have the capacity to produce and activate their own complement proteins. Thus, intrinsic renal cells express decay-accelerating factor, membrane cofactor protein, Crry, C3aR, C5aR, CR1, CR3, and CR4. These can be engaged by C3 and C5 activation products derived from systemic and local pools in renal injury. Given their capacity to provide signals that influence kidney cellular behavior, their activation can have substantial effects in renal injury. Defining these in a cell- and disease-specific fashion is an exciting challenge for future research.
Collapse
Affiliation(s)
- Tipu S Puri
- Section of Nephrology, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
32
|
Dillard P, Wetsel RA, Drouin SM. Complement C3a regulates Muc5ac expression by airway Clara cells independently of Th2 responses. Am J Respir Crit Care Med 2007; 175:1250-8. [PMID: 17400733 PMCID: PMC2176092 DOI: 10.1164/rccm.200701-049oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
RATIONALE The factors that control the secretion of epithelial mucins are essential to understanding obstructive airway diseases such as asthma. Although the complement anaphylatoxin C3a and its receptor have been shown to promote many features of allergic lung inflammation, the contribution to mucin expression has not been elucidated. OBJECTIVES To determine if the C3a receptor with its ligand regulates airway epithelial mucin production. METHODS Mice deficient in the C3a receptor were examined in a model of allergic airway disease for the presence of goblet cells and the gel-forming secreted mucin Muc5ac. MEASUREMENTS AND MAIN RESULTS Lungs from antigen-challenged C3a receptor-deficient mice revealed a dramatic decrease in goblet cells and Muc5ac compared with challenged wild-type control animals. These differences were dependent on C3a binding to its receptor since intranasal challenge with C3a induced the formation of goblet cells only in wild-type but not C3a receptor-deficient mice. Increased numbers of goblet cells were also found in C3a-stimulated RAG-1-deficient mice demonstrating a mechanism independent of T lymphocytes and Th2 cytokines, mediators which have been shown to regulate mucin expression. A direct physiological role for C3a in these models was further demonstrated in cultures of airway epithelial Clara cells, which not only express the C3a receptor but also produce Muc5ac in response to C3a. CONCLUSIONS These studies identify a novel C3a receptor-dependent mechanism in the development of airway epithelial goblet cells and regulation of Muc5ac production and implicate C3a as a mediator of airway obstruction in asthma.
Collapse
Affiliation(s)
- Patricia Dillard
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| | | | | |
Collapse
|
33
|
Dawicki W, Marshall JS. New and emerging roles for mast cells in host defence. Curr Opin Immunol 2007; 19:31-8. [PMID: 17126541 DOI: 10.1016/j.coi.2006.11.006] [Citation(s) in RCA: 208] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Accepted: 11/15/2006] [Indexed: 12/21/2022]
Abstract
Mast cells are highly effective sentinel cells, found close to blood vessels and especially common sites of potential infection, such as the skin, airways and gastrointestinal tract. Mast cells participate actively in the innate immune responses to many pathogens through a broad spectrum of mediators that can be selectively generated. They also have a role as innate effector cells in enhancing the earliest processes in the development of acquired immune responses. Studies of bacterial and parasitic models have revealed mast cell dependent regulation of effector cell recruitment, mucosal barrier function and lymph node hypertrophy. An important role for mast cells in viral infection is also implied by several in vivo and in vitro studies. There are multiple direct and indirect pathways by which mast cells can be selectively activated by pathogens including Toll-like receptors, co-receptors and complement component receptors. Understanding the mechanisms and scope of the contribution of mast cells to host defence will be crucial to regulating their activity therapeutically.
Collapse
Affiliation(s)
- Wojciech Dawicki
- Dalhousie University, 5850 College Street, Halifax, Nova Scotia, B3H 1X5, Canada
| | | |
Collapse
|
34
|
Zaidi AK, Thangam ERRB, Ali H. Distinct roles of Ca2+ mobilization and G protein usage on regulation of Toll-like receptor function in human and murine mast cells. Immunology 2007; 119:412-20. [PMID: 17067316 PMCID: PMC1819569 DOI: 10.1111/j.1365-2567.2006.02450.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Toll-like receptors (TLRs) expressed in mast cells play important roles in orchestrating host defence against bacterial pathogens. Previous studies demonstrated that TLR2 agonist tripalmitoyl-S-glycero-Cys-(Lys)4 (Pam3Cys) stimulates both degranulation and cytokine production in human mast cells but only induces cytokine production in murine mast cells. To determine the molecular basis for this difference, we utilized a human mast cell line LAD 2, murine lung and bone marrow-derived mast cells (MLMC and BMMC). We found that Pam3Cys caused a sustained Ca2+ mobilization and degranulation in LAD 2 mast cells but not in MLMC or BMMC. Despite these differences, Pam3Cys stimulated equivalent chemokine CCL2 generation in all mast cell types tested. Cyclosporin A (CsA), an inhibitor of Ca2+/calcineurin-mediated nuclear factor of activated T cells (NFAT) activation, blocked chemokine production in LAD 2 but not in MLMC or BMMC. In contrast, inhibitors of nuclear factor kappa B (NF-kappaB) completely blocked CCL2 production in MLMC and BMMC but not in LAD 2 mast cells. Pertussis toxin and U0126, which, respectively, inhibit Galphai, extracellular signal-regulated kinase (ERK) phosphorylation substantially inhibited Pam3Cys-induced CCL2 generation in LAD 2 mast cells but had little or no effect on chemokine generation in MLMC and BMMC. These findings suggest that TLR2 activation in human LAD 2 mast cells and MLMC/BMMC promotes the release of different classes of mediators via distinct signalling pathways that depend on Ca2+ mobilization and G protein usage.
Collapse
Affiliation(s)
- Asifa K Zaidi
- Department of Pathology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
35
|
El Zein N, Badran BM, Sariban E. The neuropeptide pituitary adenylate cyclase activating protein stimulates human monocytes by transactivation of the Trk/NGF pathway. Cell Signal 2007; 19:152-62. [PMID: 16914291 DOI: 10.1016/j.cellsig.2006.05.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 05/17/2006] [Accepted: 05/17/2006] [Indexed: 11/18/2022]
Abstract
Transactivation is a process whereby stimulation of G-protein-coupled receptors (GPCR) activates signaling from receptors tyrosine kinase (RTK). In neuronal cells, the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) acting through the GPCR VPAC-1 exerts trophic effects by transactivating the RTK TrkA receptor for the nerve growth factor (NGF). Both PACAP and NGF have pro-inflammatory activities on monocytes. We have tested the possibility that in monocytes, PACAP, as reported in neuronal cells, uses NGF/TrkA signaling pathway. In these cells, PACAP increases TrkA tyrosine phosphorylations through a PI-3kinase dependent but phospholipase C independent pathway. K252a, an inhibitor of TrkA decreases PACAP-induced Akt and ERK phosphorylation and calcium mobilisation resulting in decreases in intracellular H2O2 production and membrane upregulation of CD11b expression, both functions being inhibited after anti-NGF or anti-TrkA antibody treatment. K252a also inhibits PACAP-associated NF-KB activity. Monocytes increase in NGF production is seen after micromolar PACAP exposure while nanomolar treatment which desensitizes cells to high dose of PACAP prevents PACAP-induced TrkA phosphorylation, H2O2 production and CD11b expression. Finally, NGF-dependent ERK activation and H2O2 production is pertussis toxin sensitive. Altogether these data indicate that in PACAP-activated monocytes some pro-inflammatory activities occur through transactivation mechanisms involving VPAC-1, NGF and TrkA-associated tyrosine kinase activity.
Collapse
Affiliation(s)
- Nabil El Zein
- Laboratory of Pediatric Oncology, Hôpital des Enfants, 1020 Brussels, Belgium
| | | | | |
Collapse
|
36
|
Yuan Y, Fu C, Chen H, Wang X, Deng W, Huang BR. The Ran binding protein RanBPM interacts with TrkA receptor. Neurosci Lett 2006; 407:26-31. [PMID: 16959415 DOI: 10.1016/j.neulet.2006.06.059] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Revised: 06/20/2006] [Accepted: 06/28/2006] [Indexed: 10/24/2022]
Abstract
RanBPM as a novel binding protein can interact with neurotrophin receptor p75NTR and tyrosine kinase receptor Met which has a similar tyrosine kinase structure as receptor TrkA has. Whether RanBPM interacts with neurotrophin receptor TrkA has not been established to date. In this study, using yeast two-hybrid system, it was identified that RanBPM bound to the intracellular domain (ICD) of neurotrophin receptor TrkA through its SPRY motif. We confirmed the formation of complexes between RanBPM and TrkA by co-immunoprecipitation studies and GST pull-down assays. The region of TrkA interacted with the SPRY domain of RanBPM was located in its tyrosine kinase domain. Furthermore, coimmunoprecipitaiton revealed endogenous RanBPM and receptors TrkA did interact in several mammalian cell lines. It was found that the overexpression of RanBPM could inhibit NGF-induced increase of nuclear factor of activated T cells (NFAT) dependent luciferase expression through its interaction with receptor TrkA, and NFAT transcriptional activity plays an important role in neuronal signal transduction. These data suggested that RanBPM could participate in neurotrophin-mediated gene transcription and expression by its binding to TrkA.
Collapse
Affiliation(s)
- Yuhe Yuan
- National Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | | | | | | | | | | |
Collapse
|
37
|
Jha P, Sohn JH, Xu Q, Nishihori H, Wang Y, Nishihori S, Manickam B, Kaplan HJ, Bora PS, Bora NS. The complement system plays a critical role in the development of experimental autoimmune anterior uveitis. Invest Ophthalmol Vis Sci 2006; 47:1030-8. [PMID: 16505038 PMCID: PMC1975680 DOI: 10.1167/iovs.05-1062] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The role of complement in ocular autoimmunity was explored in a experimental autoimmune anterior uveitis (EAAU) animal model. METHODS EAAU was induced in Lewis rats by immunization with bovine melanin-associated antigen. Complement activation in the eye was monitored by Western blot for iC3b. The importance of complement to the development of EAAU was studied by comparing the course of intraocular inflammation in normal Lewis rats (complement-sufficient) with cobra venom factor-treated rats (complement-depleted). Eyes were harvested from both complement-sufficient and complement-depleted rats for mRNA and protein analysis for IFN-gamma, IL-10, and interferon-inducible protein (IP)-10. Intracellular adhesion molecule (ICAM)-1 and leukocyte-endothelial cell adhesion molecule (LECAM)-1 were detected by immunofluorescent staining. OX-42 was used to investigate the importance of iC3b and CR3 interaction in EAAU. RESULTS There was a correlation between ocular complement activation and disease progression in EAAU. The incidence, duration, and severity of disease were dramatically reduced after active immunization in complement-depleted rats. Complement depletion also completely suppressed adoptive transfer EAAU. The presence of complement was critical for local production of cytokines (IFN-gamma and IL-10), chemokines (IP-10), and adhesion molecules (ICAM-1 and LECAM-1) during EAAU. Furthermore, intraocular complement activation, specifically iC3b production and engagement of complement receptor 3 (CR3), had a significant impact on disease activity in EAAU. CONCLUSIONS The study provided the novel finding that complement activation plays a central role in the pathogenesis of ocular autoimmunity and may serve as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Purushottam Jha
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jeong-Hyeon Sohn
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Qin Xu
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Hiroki Nishihori
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Yali Wang
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Saori Nishihori
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Balasubramanian Manickam
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Henry J. Kaplan
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky
| | - Puran S. Bora
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nalini S. Bora
- Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
38
|
Bracci-Laudiero L, Aloe L, Caroleo MC, Buanne P, Costa N, Starace G, Lundeberg T. Endogenous NGF regulates CGRP expression in human monocytes, and affects HLA-DR and CD86 expression and IL-10 production. Blood 2005; 106:3507-14. [PMID: 16099883 DOI: 10.1182/blood-2004-10-4055] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Our recent results on autocrine nerve growth factor (NGF) synthesis in B lymphocytes, which directly regulates the expression and release of calcitonin gene-related peptide (CGRP), a neuropeptide known to down-regulate immune response, led us to propose an anti-inflammatory action of NGF. In the present work, we investigated whether the endogenous synthesis of NGF can regulate the expression of CGRP in other antigen-presenting cells, such as monocytes, and whether this may have a functional effect. Our data indicate that human monocytes synthesize basal levels of NGF and CGRP and that, following lipopolysaccharide (LPS) stimulation, NGF and CGRP expression are both up-regulated. When endogenous NGF is neutralized, the up-regulation of CGRP expression induced by LPS is inhibited. The expression of membrane molecules involved in T-cell activation such as human leukocyte antigen-DR (HLA-DR) and CD86 is affected by endogenous NGF, and similar effects were obtained using a CGRP(1) receptor antagonist. In addition, NGF deprivation in LPS-treated monocytes significantly decreases interleukin 10 (IL-10) synthesis. Our findings indicate that endogenous NGF synthesis has a functional role and may represent a physiologic mechanism to down-regulate major histocompatibility complex (MHC) class II and CD86 expression and alter the development of immune responses.
Collapse
|
39
|
Kim JY, Ro JY. Signal Pathway of Cytokines Produced by Reactive Oxygen Species Generated from Phorbol Myristate Acetate-Stimulated HMC-1 Cells. Scand J Immunol 2005; 62:25-35. [PMID: 16091123 DOI: 10.1111/j.1365-3083.2005.01636.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The relationship of cytokine production and reactive oxygen species (ROS) generated in mast cells has not been reported yet. This study aimed to examine the signal pathway in the production of cytokines [interleukin-8 (IL-8) and tumour necrosis factor-alpha (TNF-alpha)] by ROS generated from phorbol myristate acetate (PMA)-stimulated human mast cell line-1 cells (HMC-1). HMC-1 cells were stimulated with 25 ng/ml of PMA. The ROS generation and production of cytokines (IL-8 and TNF-alpha) were measured by fluorescence-activated cell sorter and enzyme-linked immunosorbent assay method, respectively. Phosphorylation of mitogen-activated protein kinase family (extracellular signal-regulated kinase, p38 and c-Jun N-terminal kinase) was detected by the Western blotting method. The expression of cytokine's mRNA was measured by reverse transcriptase--polymerase chain reaction, and the DNA-binding activity of the transcription factors [nuclear factor-kappaB (NF-kappaB) and activator protein-1] was detected by electrophoretic mobility shift assay. PMA-stimulated HMC-1 cells immediately generated ROS, and the generated ROS was inhibited by 1,3-dimethyl-2-thiourea (DMTU), but partially inhibited by catalase or N-acetyl-L-cysteine. The production of cytokines in PMA-stimulated HMC-1 cells reached the maximum at 3-5 h and was inhibited by DMTU and specific kinase inhibitor for p38, SB203580. DMTU and SB203580 also inhibited the expressed cytokine's mRNA level and the increased DNA-binding activity of transcription factors, NF-kappaB in PMA-stimulated HMC-1 cells. These data suggest that intracellular ROS generated from PMA-stimulated HMC-1 cells contributes to the production of inflammatory cytokines via p38 kinase/NF-kappaB.
Collapse
Affiliation(s)
- J Y Kim
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | | |
Collapse
|
40
|
Ali H, Panettieri RA. Anaphylatoxin C3a receptors in asthma. Respir Res 2005; 6:19. [PMID: 15723703 PMCID: PMC551592 DOI: 10.1186/1465-9921-6-19] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Accepted: 02/21/2005] [Indexed: 11/30/2022] Open
Abstract
The complement system forms the central core of innate immunity but also mediates a variety of inflammatory responses. Anaphylatoxin C3a, which is generated as a byproduct of complement activation, has long been known to activate mast cells, basophils and eosinophils and to cause smooth muscle contraction. However, the role of C3a in the pathogenesis of allergic asthma remains unclear. In this review, we examine the role of C3a in promoting asthma. Following allergen challenge, C3a is generated in the lung of subjects with asthma but not healthy subjects. Furthermore, deficiency in C3a generation or in G protein coupled receptor for C3a abrogates allergen-induced responses in murine models of pulmonary inflammation and airway hyperresponsiveness. In addition, inhibition of complement activation or administration of small molecule inhibitors of C3a receptor after sensitization but before allergen challenge inhibits airway responses. At a cellular level, C3a stimulates robust mast cell degranulation that is greatly enhanced following cell-cell contact with airway smooth muscle (ASM) cells. Therefore, C3a likely plays an important role in asthma primarily by regulating mast cell-ASM cell interaction.
Collapse
Affiliation(s)
- Hydar Ali
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 South 40Street, Philadelphia, PA, 19104, USA
| | - Reynold A Panettieri
- Pulmonary Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania, BRBII/III, 421 Curie Boulevard, Philadelphia PA 19104, USA
| |
Collapse
|