1
|
Froehlich F, Landerholm K, Neeb J, Meß AK, Seiler DL, Tilburgs T, Karsten CM. Emerging role of C5aR2: novel insights into the regulation of uterine immune cells during pregnancy. Front Immunol 2024; 15:1411315. [PMID: 38979410 PMCID: PMC11229525 DOI: 10.3389/fimmu.2024.1411315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/27/2024] [Indexed: 07/10/2024] Open
Abstract
Pregnancy is a fascinating immunological phenomenon because it allows allogeneic fetal and placental tissues to survive inside the mother. As a component of innate immunity with high inflammatory potential, the complement system must be tightly regulated during pregnancy. Dysregulation of the complement system plays a role in pregnancy complications including pre-eclampsia and intrauterine growth restriction. Complement components are also used as biomarkers for pregnancy complications. However, the mechanisms of detrimental role of complement in pregnancy is poorly understood. C5a is the most potent anaphylatoxin and generates multiple immune reactions via two transmembrane receptors, C5aR1 and C5aR2. C5aR1 is pro-inflammatory, but the role of C5aR2 remains largely elusive. Interestingly, murine NK cells have been shown to express C5aR2 without the usual co-expression of C5aR1. Furthermore, C5aR2 appears to regulate IFN-γ production by NK cells in vitro. As IFN-γ produced by uterine NK cells is one of the major factors for the successful development of a vital pregnancy, we investigated the role anaphylatoxin C5a and its receptors in the establishment of pregnancy and the regulation of uterine NK cells by examinations of murine C5ar2-/- pregnancies and human placental samples. C5ar2-/- mice have significantly reduced numbers of implantation sites and a maternal C5aR2 deficiency results in increased IL-12, IL-18 and IFN-γ mRNA expression as well as reduced uNK cell infiltration at the maternal-fetal interface. Human decidual leukocytes have similar C5a receptor expression patterns showing clinical relevance. In conclusion, this study identifies C5aR2 as a key contributor to dNK infiltration and pregnancy success.
Collapse
Affiliation(s)
- Fenna Froehlich
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Luebeck, Germany
| | - Konstanze Landerholm
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Luebeck, Germany
| | - Johanna Neeb
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Luebeck, Germany
| | - Ann-Kathrin Meß
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Luebeck, Germany
| | - Daniel Leonard Seiler
- Institute for Systemic Inflammation Research (ISEF), University of Lübeck, Luebeck, Germany
| | - Tamara Tilburgs
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | | |
Collapse
|
2
|
Lin J(C, Hwang S(W, Luo H, Mohamud Y. Double-Edged Sword: Exploring the Mitochondria-Complement Bidirectional Connection in Cellular Response and Disease. BIOLOGY 2024; 13:431. [PMID: 38927311 PMCID: PMC11200454 DOI: 10.3390/biology13060431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Mitochondria serve an ultimate purpose that seeks to balance the life and death of cells, a role that extends well beyond the tissue and organ systems to impact not only normal physiology but also the pathogenesis of diverse diseases. Theorized to have originated from ancient proto-bacteria, mitochondria share similarities with bacterial cells, including their own circular DNA, double-membrane structures, and fission dynamics. It is no surprise, then, that mitochondria interact with a bacterium-targeting immune pathway known as a complement system. The complement system is an ancient and sophisticated arm of the immune response that serves as the body's first line of defense against microbial invaders. It operates through a complex cascade of protein activations, rapidly identifying and neutralizing pathogens, and even aiding in the clearance of damaged cells and immune complexes. This dynamic system, intertwining innate and adaptive immunity, holds secrets to understanding numerous diseases. In this review, we explore the bidirectional interplay between mitochondrial dysfunction and the complement system through the release of mitochondrial damage-associated molecular patterns. Additionally, we explore several mitochondria- and complement-related diseases and the potential for new therapeutic strategies.
Collapse
Affiliation(s)
- Jingfei (Carly) Lin
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Sinwoo (Wendy) Hwang
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
3
|
Panebianco M, Ciccarese C, Strusi A, Beccia V, Carbone C, Agostini A, Piro G, Tortora G, Iacovelli R. The Role of the Complement in Clear Cell Renal Carcinoma (ccRCC)-What Future Prospects Are There for Its Use in Clinical Practice? Cancers (Basel) 2024; 16:490. [PMID: 38339243 PMCID: PMC10854780 DOI: 10.3390/cancers16030490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
In recent years, the first-line available therapeutic options for metastatic renal cell carcinoma (mRCC) have radically changed with the introduction into clinical practice of new immune checkpoint inhibitor (ICI)-based combinations. Many efforts are focusing on identifying novel prognostic and predictive markers in this setting. The complement system (CS) plays a central role in promoting the growth and progression of mRCC. In particular, mRCC has been defined as an "aggressive complement tumor", which encompasses a group of malignancies with poor prognosie and highly expressed complement components. Several preclinical and retrospective studies have demonstrated the negative prognostic role of the complement in mRCC; however, there is little evidence on its possible role as a predictor of the response to ICIs. The purpose of this review is to explore more deeply the physio-pathological role of the complement in the development of RCC and its possible future use in clinical practice as a prognostic and predictive factor.
Collapse
Affiliation(s)
- Martina Panebianco
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Chiara Ciccarese
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Alessandro Strusi
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| | - Viria Beccia
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Antonio Agostini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Geny Piro
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Giampaolo Tortora
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| | - Roberto Iacovelli
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| |
Collapse
|
4
|
Cai X, Wu J, An ZY, Wang CC, Zhu XL, He Y, Fu HX, Huang XJ, Zhang XH. Tacrolimus prevents complement-mediated Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and pyroptosis of mesenchymal stem cells from immune thrombocytopenia. Br J Haematol 2023; 202:995-1010. [PMID: 36546515 DOI: 10.1111/bjh.18625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/05/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
The abnormal immunomodulatory functions of mesenchymal stem cells (MSCs) have been implicated in the development of immune thrombocytopenia (ITP). Recent studies have suggested important effects of complement on immune cell function. However, whether complement modulates bone marrow MSCs function in ITP is poorly defined. Tacrolimus has recently been applied to the treatment of autoimmune diseases. Here, we explored whether impaired ITP-MSCs could be targeted by tacrolimus. Our results showed that the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome was activated in ITP MSCs with complement deposition (MSCs-C+ ) and initiated caspase-1-dependent pyroptosis. Transcriptome sequencing results showed abnormal fatty acid metabolism in MSCs-C+ . Enhanced fatty acid β-oxidation and reactive oxygen species production activated the NLRP3 inflammasome. Adipocytes derived from MSCs-C+ secreted less adiponectin. Adiponectin promoted the differentiation of megakaryocytes and inhibited the destruction of platelets. Tacrolimus inhibited NLRP3 inflammasome activation and MSCs-C+ pyroptosis in vitro and in vivo. Tacrolimus plus danazol elicited a higher sustained response than danazol monotherapy in corticosteroid-resistant patients with ITP. Our findings demonstrate that the activation of the NLRP3 inflammasome in ITP MSCs mediated by complement could be inhibited by tacrolimus, which might be a potential new therapy for ITP.
Collapse
Affiliation(s)
- Xuan Cai
- Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Jin Wu
- Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Zhuo-Yu An
- Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Chen-Cong Wang
- Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Lu Zhu
- Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yun He
- Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Hai-Xia Fu
- Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Beijing, China
- National Clinical Research Center for Hematologic Disease, Beijing, China
- Peking University Institute of Hematology, Beijing, China
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| |
Collapse
|
5
|
Intertwined pathways of complement activation command the pathogenesis of lupus nephritis. Transl Res 2022; 245:18-29. [PMID: 35296451 PMCID: PMC9167748 DOI: 10.1016/j.trsl.2022.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022]
Abstract
The complement system is involved in the origin of autoimmunity and systemic lupus erythematosus. Both genetic deficiency of complement components and excessive activation are involved in primary and secondary renal diseases, including lupus nephritis. Among the pathways, the classical pathway has long been accepted as the main pathway of complement activation in systemic lupus erythematosus. However, more recent studies have shown the contribution of factors B and D which implies the involvement of the alternative pathway. While there is evidence on the role of the lectin pathway in systemic lupus erythematosus, it is yet to be demonstrated whether this pathway is protective or harmful in lupus nephritis. Complement is being explored for the development of disease biomarkers and therapeutic targeting. In the current review we discuss the involvement of complement in lupus nephritis.
Collapse
|
6
|
Howell MC, Green R, McGill AR, Kahlil RM, Dutta R, Mohapatra SS, Mohapatra S. Activation of Intracellular Complement in Lungs of Patients With Severe COVID-19 Disease Decreases T-Cell Activity in the Lungs. Front Immunol 2021; 12:700705. [PMID: 34899680 PMCID: PMC8652259 DOI: 10.3389/fimmu.2021.700705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 10/25/2021] [Indexed: 12/29/2022] Open
Abstract
A novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), arose late in 2019, with disease pathology ranging from asymptomatic to severe respiratory distress with multi-organ failure requiring mechanical ventilator support. It has been found that SARS-CoV-2 infection drives intracellular complement activation in lung cells that tracks with disease severity. However, the cellular and molecular mechanisms responsible remain unclear. To shed light on the potential mechanisms, we examined publicly available RNA-Sequencing data using CIBERSORTx and conducted a Ingenuity Pathway Analysis to address this knowledge gap. In complement to these findings, we used bioinformatics tools to analyze publicly available RNA sequencing data and found that upregulation of complement may be leading to a downregulation of T-cell activity in lungs of severe COVID-19 patients. Thus, targeting treatments aimed at the modulation of classical complement and T-cell activity may help alleviate the proinflammatory effects of COVID-19, reduce lung pathology, and increase the survival of COVID-19 patients.
Collapse
Affiliation(s)
- Mark C Howell
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida (USF), Tampa, FL, United States.,James A. Haley Veterans Hospital, Department of Veterans Affairs, Tampa, FL, United States
| | - Ryan Green
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida (USF), Tampa, FL, United States.,James A. Haley Veterans Hospital, Department of Veterans Affairs, Tampa, FL, United States
| | - Andrew R McGill
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida (USF), Tampa, FL, United States.,James A. Haley Veterans Hospital, Department of Veterans Affairs, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida (USF), Tampa, FL, United States
| | - Roukiah M Kahlil
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida (USF), Tampa, FL, United States
| | - Rinku Dutta
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida (USF), Tampa, FL, United States
| | - Shyam S Mohapatra
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida (USF), Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida (USF), Tampa, FL, United States
| | - Subhra Mohapatra
- James A. Haley Veterans Hospital, Department of Veterans Affairs, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida (USF), Tampa, FL, United States
| |
Collapse
|
7
|
Liu S, Liu F, Wang T, Liu J, Hu C, Sun L, Wang G. Polysaccharides Extracted From Panax Ginseng C.A. Mey Enhance Complement Component 4 Biosynthesis in Human Hepatocytes. Front Pharmacol 2021; 12:734394. [PMID: 34566655 PMCID: PMC8461058 DOI: 10.3389/fphar.2021.734394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/30/2021] [Indexed: 11/23/2022] Open
Abstract
Panax ginseng C.A. Mey (ginseng) is a classic medicinal plant which is well known for enhancing immune capacity. Polysaccharides are one of the main active components of ginseng. We isolated water-soluble ginseng polysaccharides (WGP) and analyzed the physicochemical properties of WGP including molecular weight, monosaccharide composition, and structural characteristics. WGP had minimal effect on the growth of hepatocytes. Interestingly, WGP significantly increased the mRNA and protein levels of complement component 4 (C4), one of the core components of the complement system. Promoter reporter gene assays revealed that WGP significantly enhanced activity of the C4 gene promoter. Deletion analyses determined that the E-box1 and Sp1 regions play key roles in WGP-induced C4 transcription. Taken together, our results suggest that WGP promotes C4 biosynthesis through upregulation of transcription. These results provide new explanation for the intrinsic mechanism by which ginseng boosts human immune capacity.
Collapse
Affiliation(s)
- Shuang Liu
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, China
| | - Fangbing Liu
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, China
| | - Tingting Wang
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, China
| | - Jianzeng Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Cheng Hu
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital of Changchun University of Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Changchun University of Chinese Medicine, Changchun, China
| | - Guan Wang
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
8
|
Kolev M, West EE, Kunz N, Chauss D, Moseman EA, Rahman J, Freiwald T, Balmer ML, Lötscher J, Dimeloe S, Rosser EC, Wedderburn LR, Mayer-Barber KD, Bohrer A, Lavender P, Cope A, Wang L, Kaplan MJ, Moutsopoulos NM, McGavern D, Holland SM, Hess C, Kazemian M, Afzali B, Kemper C. Diapedesis-Induced Integrin Signaling via LFA-1 Facilitates Tissue Immunity by Inducing Intrinsic Complement C3 Expression in Immune Cells. Immunity 2020; 52:513-527.e8. [PMID: 32187519 DOI: 10.1016/j.immuni.2020.02.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 12/30/2019] [Accepted: 02/19/2020] [Indexed: 12/12/2022]
Abstract
Intrinsic complement C3 activity is integral to human T helper type 1 (Th1) and cytotoxic T cell responses. Increased or decreased intracellular C3 results in autoimmunity and infections, respectively. The mechanisms regulating intracellular C3 expression remain undefined. We identified complement, including C3, as among the most significantly enriched biological pathway in tissue-occupying cells. We generated C3-reporter mice and confirmed that C3 expression was a defining feature of tissue-immune cells, including T cells and monocytes, occurred during transendothelial diapedesis, and depended on integrin lymphocyte-function-associated antigen 1 (LFA-1) signals. Immune cells from patients with leukocyte adhesion deficiency type 1 (LAD-1) had reduced C3 transcripts and diminished effector activities, which could be rescued proportionally by intracellular C3 provision. Conversely, increased C3 expression by T cells from arthritis patients correlated with disease severity. Our study defines integrins as key controllers of intracellular complement, demonstrates that perturbations in the LFA-1-C3-axis contribute to primary immunodeficiency, and identifies intracellular C3 as biomarker of severity in autoimmunity.
Collapse
Affiliation(s)
- Martin Kolev
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Erin E West
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Natalia Kunz
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Daniel Chauss
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD 20892, USA
| | - E Ashley Moseman
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Jubayer Rahman
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Tilo Freiwald
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD 20892, USA
| | - Maria L Balmer
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Jonas Lötscher
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Sarah Dimeloe
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel 4031, Switzerland; Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Elizabeth C Rosser
- Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; Arthritis Research UK Centre for Adolescent Rheumatology at UCL, UCHL and GOSH, London WC1N 1EH, UK
| | - Lucy R Wedderburn
- Infection, Immunity, Inflammation Programme, University College London (UCL) Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; Arthritis Research UK Centre for Adolescent Rheumatology at UCL, UCHL and GOSH, London WC1N 1EH, UK; National Institute for Health Research (NIHR) Biomedical Research Centre at Great Ormond Street NHS Foundation Trust, London WC1N 1EH, UK
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Andrea Bohrer
- Inflammation and Innate Immunity Unit, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD 20892, USA
| | - Paul Lavender
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, UK
| | - Andrew Cope
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, UK
| | - Luopin Wang
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN 47907, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Disease (NIAMS), NIH, Bethesda, MD 20892, USA
| | - Niki M Moutsopoulos
- Oral Immunity and Inflammation Unit, National Institute of Dental and Craniofacial Research (NIDCR), NIH, Bethesda, MD 20892, USA
| | - Dorian McGavern
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD 20892, USA
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Christoph Hess
- Department of Biomedicine, Immunobiology, University Hospital and University of Basel, Basel 4031, Switzerland; Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD 20892, USA.
| | - Claudia Kemper
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, UK; Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany.
| |
Collapse
|
9
|
Wynne M, Atkinson C, Schlosser RJ, Mulligan JK. Contribution of Epithelial Cell Dysfunction to the Pathogenesis of Chronic Rhinosinusitis with Nasal Polyps. Am J Rhinol Allergy 2019; 33:782-790. [PMID: 31382760 DOI: 10.1177/1945892419868588] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background In the past, the airway epithelium was thought to be primarily an inert physical barrier. We now know that the upper airway epithelium plays a critical role in both innate and adaptive immunity, and that epithelial dysfunction is strongly associated with inflammatory airway disease. The pathogenesis of chronic rhinosinusitis is poorly understood, but growing evidence supports a key role for the airway epithelium in the pathophysiology of the disease. Objective The purpose of this study is to explore our current understanding of how dysfunction in human sinonasal epithelial cells (HSNECs) contributes to the pathogenesis of chronic rhinosinusitis with nasal polyps (CRSwNP) and to examine how current and developing therapies affect epithelial cell functions. Methods A literature review of papers published in English pertaining to epithelial cell dysfunction in patients with CRSwNP was performed using the PubMed database. The search utilized combinations of the following key words: sinusitis, polyps, epithelium, pathophysiology, barrier function, dendritic cells, eosinophils, T cells, complement, mucociliary clearance, vitamin D, cytokines, chemokines, taste receptors, steroids, saline, and therapy. Results HSNEC mucociliary clearance, barrier function, secretion of cytokines, influence on dendritic cells, influence on T-cells, regulation of eosinophils, vitamin D metabolism, complement production, and taste receptor function are altered in patients with CRSwNP and contribute to the pathogenesis of the disease. Current therapies utilized to manage CRSwNP counteract the effects of HSNEC dysfunction and relieve key symptoms of the disease. Conclusion HSNECs are key players in both innate and adaptive immunity, and altered epithelial functions are closely intertwined with the pathogenesis of CRSwNP. Our review supports further investigation of altered HSNEC function in patients with CRSwNP and supports development of novel epithelial-targeted therapies for its management.
Collapse
Affiliation(s)
- Michael Wynne
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Department of Surgery, Medical University of South Carolina, Lee Patterson Allen Transplant Immunobiology Laboratory, Charleston, South Carolina
| | - Rodney J Schlosser
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Jennifer K Mulligan
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina.,Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
10
|
Satyam A, Graef ER, Lapchak PH, Tsokos MG, Dalle Lucca JJ, Tsokos GC. Complement and coagulation cascades in trauma. Acute Med Surg 2019; 6:329-335. [PMID: 31592318 PMCID: PMC6773636 DOI: 10.1002/ams2.426] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 03/19/2019] [Indexed: 01/12/2023] Open
Abstract
Trauma remains a major cause of death throughout the world, especially for patients younger than 45 years. Due to rapid advances in clinical management, both in the acute and prehospital settings, trauma patients survive devastating injuries at unprecedented rates. However, these patients can often face life threatening complications that stem from the robust innate immune response induced by severe hemorrhage, leading to further tissue injury rather than repair. The complement and coagulation cascades are key mediators in this disordered reaction, which includes the development of trauma‐induced coagulopathy. There is increasing evidence that cross‐talk between these two pathways allows rapid amplification of their otherwise targeted responses and contributes to overwhelming and prolonged systemic inflammation. In this article, we summarize the initial steps of innate immune response to trauma and review the complex complement and coagulation cascades, as well as how they interact with each other. Despite progress in understanding these cascades, effective therapeutic targets have yet to be found and further research is needed both to improve survival rates as well as decrease associated morbidity.
Collapse
Affiliation(s)
- Abhigyan Satyam
- Department of Medicine Beth Israel Deaconess Medical Center Harvard Medical School Boston Massachusetts
| | - Elizabeth R Graef
- Department of Medicine Beth Israel Deaconess Medical Center Harvard Medical School Boston Massachusetts
| | - Peter H Lapchak
- Department of Medicine Beth Israel Deaconess Medical Center Harvard Medical School Boston Massachusetts
| | - Maria G Tsokos
- Department of Medicine Beth Israel Deaconess Medical Center Harvard Medical School Boston Massachusetts
| | | | - George C Tsokos
- Department of Medicine Beth Israel Deaconess Medical Center Harvard Medical School Boston Massachusetts
| |
Collapse
|
11
|
Hansen CB, Willer A, Bayarri-Olmos R, Kemper C, Garred P. Expression of complement C3, C5, C3aR and C5aR1 genes in resting and activated CD4 + T cells. Immunobiology 2018; 224:307-315. [PMID: 30612786 DOI: 10.1016/j.imbio.2018.12.004] [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: 11/16/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 01/02/2023]
Abstract
Complement activation is traditionally thought to occur in the extracellular space. However, it has been suggested that complement proteins are activated and function at additional locations. T cells contain intracellular stores of C3 and C5 that can be cleaved into C3a and C5a and bind to intracellular receptors, which have been shown to be of vital importance for the differentiation and function of these cells. However, whether the origin of the complement proteins located within T cells is derived from endogenous produced complement or from an uptake dependent mechanism is unknown. The presence of intracellular C3 in T cells from normal donors was investigated by fluorescence microscopy and flow cytometry. Moreover, mRNA expression levels of several genes encoding for complement proteins with primary focus on C3, C3aR, C5 and C5aR1 during resting state and upon activation of CD4+ T cells were investigated by a quantitative PCR technique. Furthermore, the gene expression level was evaluated at different time points. We confirmed the presence of intracellular C3 protein in normal T-cells. However, we could not see any increase in mRNA levels using any activation strategy tested. On the contrary, we observed a slight increase in C3 and C5aR1 mRNA only in the non-activated T-cells compared to the activated T cells, and a decrease in the activated T-cells at different incubation time points. Our results show that there is a baseline intracellular expression of the complement C3, C5, C3aR and C5aR1 genes in normal CD4+ T cells, but that expression is not increased during T-cell activation, but rather down regulated. Thus, the pool of intracellular complement in CD4+ T cells may either be due to accumulated complement due low-grade expression or arise from the circulation from an uptake dependent mechanism, but these possibilities are not mutually exclusive.
Collapse
Affiliation(s)
- Cecilie Bo Hansen
- Laboratory of Molecular Medicine, Department of Clinical Immunology Section 7631, Faculty of Health and Medical Sciences, University Hospital of Copenhagen, Denmark
| | - Anton Willer
- Laboratory of Molecular Medicine, Department of Clinical Immunology Section 7631, Faculty of Health and Medical Sciences, University Hospital of Copenhagen, Denmark
| | - Rafael Bayarri-Olmos
- Laboratory of Molecular Medicine, Department of Clinical Immunology Section 7631, Faculty of Health and Medical Sciences, University Hospital of Copenhagen, Denmark
| | - Claudia Kemper
- National Heart, Lung and Blood Institute, National Institute of Health, Bethesda, MD, 20814, USA
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology Section 7631, Faculty of Health and Medical Sciences, University Hospital of Copenhagen, Denmark.
| |
Collapse
|
12
|
Arbore G, West EE, Rahman J, Le Friec G, Niyonzima N, Pirooznia M, Tunc I, Pavlidis P, Powell N, Li Y, Liu P, Servais A, Couzi L, Fremeaux-Bacchi V, Placais L, Ferraro A, Walsh PR, Kavanagh D, Afzali B, Lavender P, Lachmann HJ, Kemper C. Complement receptor CD46 co-stimulates optimal human CD8 + T cell effector function via fatty acid metabolism. Nat Commun 2018; 9:4186. [PMID: 30305631 PMCID: PMC6180132 DOI: 10.1038/s41467-018-06706-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022] Open
Abstract
The induction of human CD4+ Th1 cells requires autocrine stimulation of the complement receptor CD46 in direct crosstalk with a CD4+ T cell-intrinsic NLRP3 inflammasome. However, it is unclear whether human cytotoxic CD8+ T cell (CTL) responses also rely on an intrinsic complement-inflammasome axis. Here we show, using CTLs from patients with CD46 deficiency or with constitutively-active NLRP3, that CD46 delivers co-stimulatory signals for optimal CTL activity by augmenting nutrient-influx and fatty acid synthesis. Surprisingly, although CTLs express NLRP3, a canonical NLRP3 inflammasome is not required for normal human CTL activity, as CTLs from patients with hyperactive NLRP3 activity function normally. These findings establish autocrine complement and CD46 activity as integral components of normal human CTL biology, and, since CD46 is only present in humans, emphasize the divergent roles of innate immune sensors between mice and men.
Collapse
Affiliation(s)
- Giuseppina Arbore
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milano, Italy
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Erin E West
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jubayer Rahman
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Gaelle Le Friec
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Nathalie Niyonzima
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mehdi Pirooznia
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Ilker Tunc
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Nicholas Powell
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Yuesheng Li
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Poching Liu
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Aude Servais
- Service de Néphrologie adulte, Hôpital Necker, Paris, France
| | - Lionel Couzi
- Nephrologie,Transplantation, Dialyse, CHU Bordeaux, and CNRS-UMR 5164 Immuno ConcEpT, Université de Bordeaux, Bordeaux, France
| | - Veronique Fremeaux-Bacchi
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, and INSERM UMR S1138, Centre de Recherche des Cordeliers, Paris, France
| | - Leo Placais
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Alastair Ferraro
- Department of Renal Medicine, Nottingham University Hospitals, NHS Trust, Nottingham, UK
| | - Patrick R Walsh
- National Renal Complement Therapeutics Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - David Kavanagh
- National Renal Complement Therapeutics Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Behdad Afzali
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
- Immunoregulation Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Paul Lavender
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Helen J Lachmann
- UK National Amyloidosis Centre, Division of Medicine, University College London, Royal Free Campus, London, UK.
| | - Claudia Kemper
- School of Immunology and Microbial Sciences, King's College London, London, UK.
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
13
|
Mulligan JK, Patel K, Williamson T, Reaves N, Carroll W, Stephenson SE, Gao P, Drake RR, Neely BA, Tomlinson S, Schlosser RJ, Atkinson C. C3a receptor antagonism as a novel therapeutic target for chronic rhinosinusitis. Mucosal Immunol 2018; 11:1375-1385. [PMID: 29907871 PMCID: PMC6162114 DOI: 10.1038/s41385-018-0048-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 04/05/2018] [Accepted: 04/30/2018] [Indexed: 02/04/2023]
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) is an inflammatory disease with an unknown etiology. Recent studies have implicated the complement system as a potential modulator of disease immunopathology. We performed proteomic pathway enrichment analysis of differentially increased proteins, and found an enrichment of complement cascade pathways in the nasal mucus of individuals with CRSwNP as compared to control subjects. Sinonasal mucus levels of complement 3 (C3) correlated with worse subjective disease severity, whereas no significant difference in systemic C3 levels could be determined in plasma samples. Given that human sinonasal epithelial cells were the predominate sinonasal source of C3 and complement anaphylatoxin 3a (C3a) staining, we focused on their role in in vitro studies. Baseline intracellular C3 levels were higher in CRSwNP cells, and following exposure to Aspergillus fumigatus (Af) extract, they released significantly more C3 and C3a. Inhibition of complement 3a receptor (C3aR) signaling led to a decrease in Af-induced C3 and C3a release, both in vitro and in vivo. Finally, we found in vivo that C3aR deficiency or inhibition significantly reduced inflammation and CRS development in a mouse model of Af-induced CRS. These findings demonstrate that local sinonasal complement activation correlates with subjective disease severity, and that local C3aR antagonism significantly ameliorates Af-induced CRS in a rodent model.
Collapse
Affiliation(s)
- Jennifer K Mulligan
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Kunal Patel
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Lee Patterson Allen Transplant Immunobiology Laboratory, Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Tucker Williamson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Nicholas Reaves
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - William Carroll
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Sarah E Stephenson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Peng Gao
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Benjamin A Neely
- Marine Biochemical Sciences, National Institute of Standards and Technology, Charleston, SC, USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Rodney J Schlosser
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Lee Patterson Allen Transplant Immunobiology Laboratory, Department of Surgery, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
14
|
Abstract
The complement system is an evolutionarily ancient key component of innate immunity required for the detection and removal of invading pathogens. It was discovered more than 100 years ago and was originally defined as a liver-derived, blood-circulating sentinel system that classically mediates the opsonization and lytic killing of dangerous microbes and the initiation of the general inflammatory reaction. More recently, complement has also emerged as a critical player in adaptive immunity via its ability to instruct both B and T cell responses. In particular, work on the impact of complement on T cell responses led to the surprising discoveries that the complement system also functions within cells and is involved in regulating basic cellular processes, predominantly those of metabolic nature. Here, we review current knowledge about complement's role in T cell biology, with a focus on the novel intracellular and noncanonical activities of this ancient system.
Collapse
Affiliation(s)
- Erin E West
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, Maryland 20892, United States; ,
| | - Martin Kolev
- Division of Transplant Immunology and Mucosal Biology, King's College London, London SE1 9RT, United Kingdom;
| | - Claudia Kemper
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, Maryland 20892, United States; ,
- Division of Transplant Immunology and Mucosal Biology, King's College London, London SE1 9RT, United Kingdom;
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
15
|
Hornum L, Hansen AJ, Tornehave D, Fjording MS, Colmenero P, Wätjen IF, Søe Nielsen NH, Bliddal H, Bartels EM. C5a and C5aR are elevated in joints of rheumatoid and psoriatic arthritis patients, and C5aR blockade attenuates leukocyte migration to synovial fluid. PLoS One 2017; 12:e0189017. [PMID: 29220376 PMCID: PMC5722346 DOI: 10.1371/journal.pone.0189017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/18/2017] [Indexed: 11/17/2022] Open
Abstract
Complement activation correlates to rheumatoid arthritis disease activity, and increased amounts of the complement split product C5a is observed in synovial fluids from rheumatoid arthritis patients. Blockade of C5a or its receptor (C5aR) is efficacious in several arthritis models. The aim of this study was to investigate the role of C5a and C5aR in human rheumatoid arthritis and psoriatic arthritis–both with respect to expression and function. Synovial fluid, blood and synovial samples were obtained from rheumatoid arthritis, psoriatic arthritis and osteoarthritis patients as a less inflammatory arthritis type, and blood from healthy subjects. Cells infiltrating synovial tissue were analysed by immunohistochemistry and flow cytometry. SF and blood were analysed for biomarkers by flow cytometry or ELISA. The effect of a blocking anti-human C5aR mAb on leukocyte migration was determined using a Boyden chamber. Appropriate statistical tests were applied for comparisons. C5aR+ cells were detected in most rheumatoid arthritis, in all psoriatic arthritis, but not in non-inflammatory control synovia. C5aR+ cells were primarily neutrophils and macrophages. C5aR+ macrophages were mainly found in lymphoid aggregates in close contact with T cells. C5a levels were increased in both rheumatoid arthritis and psoriatic arthritis synovial fluid compared to osteoarthritis, and in blood from rheumatoid arthritis compared to healthy subjects. Neutrophil and monocyte migration to rheumatoid arthritis synovial fluid was significantly inhibited by anti-C5aR. The data support that the C5a-C5aR axis may be driving the infiltration of inflammatory cells into the synovial fluid and synovium in both rheumatoid and psoriatic arthritis, and suggest that C5a or C5aR may be a promising treatment target in both diseases.
Collapse
Affiliation(s)
| | | | | | | | - Paula Colmenero
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Inger Falbe Wätjen
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Frederiksberg, Denmark
| | | | - Henning Bliddal
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Frederiksberg, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Else Marie Bartels
- The Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Frederiksberg, Denmark
| |
Collapse
|
16
|
Berraondo P, Minute L, Ajona D, Corrales L, Melero I, Pio R. Innate immune mediators in cancer: between defense and resistance. Immunol Rev 2017; 274:290-306. [PMID: 27782320 DOI: 10.1111/imr.12464] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic inflammation in the tumor microenvironment and evasion of the antitumor effector immune response are two of the emerging hallmarks required for oncogenesis and cancer progression. The innate immune system not only plays a critical role in perpetuating these tumor-promoting hallmarks but also in developing antitumor adaptive immune responses. Thus, understanding the dual role of the innate system in cancer immunology is required for the design of combined immunotherapy strategies able to tackle established tumors. Here, we review recent advances in the understanding of the role of cell populations and soluble components of the innate immune system in cancer, with a focus on complement, the adapter molecule Stimulator of Interferon Genes, natural killer cells, myeloid cells, and B cells.
Collapse
Affiliation(s)
- Pedro Berraondo
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Luna Minute
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Daniel Ajona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Solid Tumors and Biomarkers, CIMA, Pamplona, Spain.,Deparment of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | | | - Ignacio Melero
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Ruben Pio
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain. .,Program of Solid Tumors and Biomarkers, CIMA, Pamplona, Spain. .,Deparment of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| |
Collapse
|
17
|
Freeley S, Kemper C, Le Friec G. The "ins and outs" of complement-driven immune responses. Immunol Rev 2017; 274:16-32. [PMID: 27782335 DOI: 10.1111/imr.12472] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The complement system represents an evolutionary old and critical component of innate immunity where it forms the first line of defense against invading pathogens. Originally described as a heat-labile fraction of the serum responsible for the opsonization and subsequent lytic killing of bacteria, work over the last century firmly established complement as a key mediator of the general inflammatory response but also as an acknowledged vital bridge between innate and adaptive immunity. However, recent studies particularly spanning the last decade have provided new insights into the novel modes and locations of complement activation and highlighted unexpected additional biological functions for this ancient system, for example, in regulating basic processes of the cell. In this review, we will cover the current knowledge about complement's established and novel roles in innate and adaptive immunity with a focus on the functional differences between serum circulating and intracellularly active complement and will describe and discuss the newly discovered cross-talks of complement with other cell effector systems particularly during T-cell induction and contraction.
Collapse
Affiliation(s)
- Simon Freeley
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK. .,Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Gaëlle Le Friec
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
18
|
van Leeuwen-Kerkhoff N, Lundberg K, Westers TM, Kordasti S, Bontkes HJ, de Gruijl TD, Lindstedt M, van de Loosdrecht AA. Transcriptional profiling reveals functional dichotomy between human slan + non-classical monocytes and myeloid dendritic cells. J Leukoc Biol 2017; 102:1055-1068. [PMID: 28720687 DOI: 10.1189/jlb.3ma0117-037r] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/31/2017] [Accepted: 06/20/2017] [Indexed: 11/24/2022] Open
Abstract
Human 6-sulfo LacNac-positive (slan+) cells have been subject to a paradigm debate. They have previously been classified as a distinct dendritic cell (DC) subset. However, evidence has emerged that they may be more related to monocytes than to DCs. To gain deeper insight into the functional specialization of slan+ cells, we have compared them with both conventional myeloid DC subsets (CD1c+ and CD141+) in human peripheral blood (PB). With the use of genome-wide transcriptional profiling, as well as functional tests, we clearly show that slan+ cells form a distinct, non-DC-like population. They cluster away from both DC subsets, and their gene-expression profile evidently suggests involvement in distinct inflammatory processes. An extensive transcriptional meta-analysis confirmed the relationship of slan+ cells with the monocytic compartment rather than with DCs. From a functional perspective, their ability to prime CD4+ and CD8+ T cells is relatively low. Combined with the finding that "antigen presentation by MHC class II" is at the top of under-represented pathways in slan+ cells, this points to a minimal role in directing adaptive T cell immunity. Rather, the higher expression levels of complement receptors on their cell surface, together with their high secretion of IL-1β and IL-6, imply a specific role in innate inflammatory processes, which is consistent with their recent identification as non-classical monocytes. This study extends our knowledge on DC/monocyte subset biology under steady-state conditions and contributes to our understanding of their role in immune-mediated diseases and their potential use in immunotherapeutic strategies.
Collapse
Affiliation(s)
- Nathalie van Leeuwen-Kerkhoff
- Department of Hematology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Theresia M Westers
- Department of Hematology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Shahram Kordasti
- Department of Haematological Medicine, King's College London and King's College Hospital, London, United Kingdom
| | - Hetty J Bontkes
- Department of Oral Cell Biology, Academic Center for Dentistry, Amsterdam, The Netherlands; and
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Arjan A van de Loosdrecht
- Department of Hematology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam University Medical Center, Amsterdam, The Netherlands;
| |
Collapse
|
19
|
Busch C, Annamalai B, Abdusalamova K, Reichhart N, Huber C, Lin Y, Jo EAH, Zipfel PF, Skerka C, Wildner G, Diedrichs-Möhring M, Rohrer B, Strauß O. Anaphylatoxins Activate Ca 2+, Akt/PI3-Kinase, and FOXO1/FoxP3 in the Retinal Pigment Epithelium. Front Immunol 2017; 8:703. [PMID: 28663750 PMCID: PMC5472091 DOI: 10.3389/fimmu.2017.00703] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/31/2017] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The retinal pigment epithelium (RPE) is a main target for complement activation in age-related macular degeneration (AMD). The anaphylatoxins C3a and C5a have been thought to mostly play a role as chemoattractants for macrophages and immune cells; here, we explore whether they trigger RPE alterations. Specifically, we investigated the RPE as a potential immunoregulatory gate, allowing for active changes in the RPE microenvironment in response to complement. DESIGN In vitro and in vivo analysis of signaling pathways. METHODS Individual activities of and interaction between the two anaphylatoxin receptors were tested in cultured RPE cells by fluorescence microscopy, western blot, and immunohistochemistry. MAIN OUTCOME MEASURES Intracellular free calcium, protein phosphorylation, immunostaining of tissues/cells, and multiplex secretion assay. RESULTS Similar to immune cells, anaphylatoxin exposure resulted in increases in free cytosolic Ca2+, PI3-kinase/Akt activation, FoxP3 and FOXO1 phosphorylation, and cytokine/chemokine secretion. Differential responses were elicited depending on whether C3a and C5a were co-administered or applied consecutively, and response amplitudes in co-administration experiments ranged from additive to driven by C5a (C3a + C5a = C5a) or being smaller than those elicited by C3a alone (C3a + C5a < C3a). CONCLUSION We suggest that this combination of integrative signaling between C3aR and C5aR helps the RPE to precisely adopt its immune regulatory function. These data further contribute to our understanding of AMD pathophysiology.
Collapse
Affiliation(s)
- Catharina Busch
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | | | - Khava Abdusalamova
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
| | - Nadine Reichhart
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
| | - Christian Huber
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
- Department of Ophthalmology, University of Heidelberg, Heidelberg, Germany
| | - Yuchen Lin
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Emeraldo A. H. Jo
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Peter F. Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Gerhild Wildner
- Department of Ophthalmology, Section of Immunobiology, Clinic of the LMU Munich, Munich, Germany
| | - Maria Diedrichs-Möhring
- Department of Ophthalmology, Section of Immunobiology, Clinic of the LMU Munich, Munich, Germany
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, United States
- Ralph H. Johnson VA Medical Center, Division of Research, Charleston, SC, United States
| | - Olaf Strauß
- Department of Ophthalmology, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
20
|
Attenuation of cGVHD by C5a/C5aR blockade is associated with increased frequency of Treg. Sci Rep 2017; 7:3603. [PMID: 28620195 PMCID: PMC5472632 DOI: 10.1038/s41598-017-03700-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 05/18/2017] [Indexed: 12/29/2022] Open
Abstract
C5aR signaling plays an important role in the regulation of T cell activation and alloimmune responses in chronic graft-versus-host disease (cGVHD). However, direct evidence of this modulation and the efficacy of C5aR blockade in the treatment of cGVHD have not been demonstrated. We observed higher expression of C5aR on both monocytes and T cells of patients with cGVHD compared with healthy controls and non-GVHD patients after allogeneic hematopoietic stem cell transplantation. Our data also demonstrated a significant negative correlation between C5aR expression and regulatory T cells (Treg) frequency in cGVHD patients, indicating a potential role of C5aR in the generation and regulation of Treg. In addition, an in vitro experiment revealed C5aR deficiency promoted the development of Treg whereas C5a activation abolished the differentiation of Treg. Importantly, we found C5aR blockade by PMX53 attenuated the pathology of cGVHD and improved the survival of cGVHD mice. PMX53 had a direct regulatory effect on Treg commitment and increased TGF-β1 expression. Thus, C5aR signaling may induce and intensify cGVHD by down-regulating Treg induction. The modulation of C5aR activation by PMX53 may provide a potential therapy for cGVHD.
Collapse
|
21
|
Intracellular complement - the complosome - in immune cell regulation. Mol Immunol 2017; 89:2-9. [PMID: 28601357 PMCID: PMC7112704 DOI: 10.1016/j.molimm.2017.05.012] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 05/14/2017] [Accepted: 05/19/2017] [Indexed: 12/18/2022]
Abstract
The complement system was defined over a century ago based on its ability to "complement" the antibody-mediated and cell-mediated immune responses against pathogens. Today our understanding of this ancient part of innate immunity has changed substantially and we know now that complement plays an undisputed pivotal role in the regulation of both innate and adaptive immunity. The complement system consists of over 50 blood-circulating, cell-surface expressed and intracellular proteins. It is key in the recognition and elimination of invading pathogens, also in the removal of self-derived danger such as apoptotic cells, and it supports innate immune responses and the initiation of the general inflammatory reactions. The long prevailing classic view of complement was that of a serum-operative danger sensor and first line of defence system, however, recent experimental and clinical evidences have demonstrated that "local" tissue and surprisingly intracellular complement (the complosome) activation impacts on normal cell physiology. This review will focus on novel aspects of intracellular complement activation and its unexpected roles in basic cell processes such as metabolism. We also discuss what the existence of the complosome potentially means for how the host handles intracellular pathogens such as viruses.
Collapse
|
22
|
Lubbers R, van Essen MF, van Kooten C, Trouw LA. Production of complement components by cells of the immune system. Clin Exp Immunol 2017; 188:183-194. [PMID: 28249350 DOI: 10.1111/cei.12952] [Citation(s) in RCA: 320] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2017] [Indexed: 12/14/2022] Open
Abstract
The complement system is an important part of the innate immune defence. It contributes not only to local inflammation, removal and killing of pathogens, but it also assists in shaping of the adaptive immune response. Besides a role in inflammation, complement is also involved in physiological processes such as waste disposal and developmental programmes. The complement system comprises several soluble and membrane-bound proteins. The bulk of the soluble proteins is produced mainly by the liver. While several complement proteins are produced by a wide variety of cell types, other complement proteins are produced by only a few related cell types. As these data suggest that local production by specific cell types may have specific functions, more detailed studies have been employed recently analysing the local and even intracellular role of these complement proteins. Here we review the current knowledge about extrahepatic production and/or secretion of complement components. More specifically, we address what is known about complement synthesis by cells of the human immune system.
Collapse
Affiliation(s)
- R Lubbers
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | - M F van Essen
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - C van Kooten
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - L A Trouw
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
23
|
Liszewski MK, Elvington M, Kulkarni HS, Atkinson JP. Complement's hidden arsenal: New insights and novel functions inside the cell. Mol Immunol 2017; 84:2-9. [PMID: 28196665 DOI: 10.1016/j.molimm.2017.01.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/07/2017] [Indexed: 12/31/2022]
Abstract
A key component of both innate and adaptive immunity, new understandings of the complement system are expanding its roles beyond that traditionally appreciated. Evidence is accumulating that complement has an intracellular arsenal of components that provide not only immune defense, but also assist in key interactions for host cell functions. Although early work has primarily centered on T cells, the intracellular complement system likely functions in many if not most cells of the body. Some of these functions may trace their origins to the primitive complement system that began as a primeval form of C3 likely tasked for protection from intracellular pathogen invasion. This later expanded to include extracellular defense as C3 became a secreted protein to patrol the vasculature. Other components were added to the growing system including regulators to protect host cells from the indiscriminate effects of this potent system. Contemporary cells may retain some of these vestigial remnants. We now know that a) C3 serves as a damage-associated molecular pattern (in particular by coating pathogens that translocate into cells), b) most cells store C3 and recycle C3(H2O) for immediate use, and c) C3 assists in cellular survival and metabolic reprogramming. Other components also are part of this hidden arsenal including C5, properdin, factors H and B, and complement receptors. Importantly, better definition of the intracellular complement system may translate into new target discovery to assist in creating the next generation of complement therapeutics.
Collapse
Affiliation(s)
| | | | - Hrishikesh S Kulkarni
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Campus Box 8045, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | |
Collapse
|
24
|
Kolev M, Kemper C. Keeping It All Going-Complement Meets Metabolism. Front Immunol 2017; 8:1. [PMID: 28149297 PMCID: PMC5241319 DOI: 10.3389/fimmu.2017.00001] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/03/2017] [Indexed: 01/22/2023] Open
Abstract
The complement system is an evolutionary old and crucial component of innate immunity, which is key to the detection and removal of invading pathogens. It was initially discovered as a liver-derived sentinel system circulating in serum, the lymph, and interstitial fluids that mediate the opsonization and lytic killing of bacteria, fungi, and viruses and the initiation of the general inflammatory responses. Although work performed specifically in the last five decades identified complement also as a critical instructor of adaptive immunity—indicating that complement’s function is likely broader than initially anticipated—the dominant opinion among researchers and clinicians was that the key complement functions were in principle defined. However, there is now a growing realization that complement activity goes well beyond “classic” immune functions and that this system is also required for normal (neuronal) development and activity and general cell and tissue integrity and homeostasis. Furthermore, the recent discovery that complement activation is not confined to the extracellular space but occurs within cells led to the surprising understanding that complement is involved in the regulation of basic processes of the cell, particularly those of metabolic nature—mostly via novel crosstalks between complement and intracellular sensor, and effector, pathways that had been overlooked because of their spatial separation. These paradigm shifts in the field led to a renaissance in complement research and provide new platforms to now better understand the molecular pathways underlying the wide-reaching effects of complement functions in immunity and beyond. In this review, we will cover the current knowledge about complement’s emerging relationship with the cellular metabolism machinery with a focus on the functional differences between serum-circulating versus intracellularly active complement during normal cell survival and induction of effector functions. We will also discuss how taking a closer look into the evolution of key complement components not only made the functional connection between complement and metabolism rather “predictable” but how it may also give clues for the discovery of additional roles for complement in basic cellular processes.
Collapse
Affiliation(s)
- Martin Kolev
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK; Laboratory of Molecular Immunology, The Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
25
|
Hovland A, Jonasson L, Garred P, Yndestad A, Aukrust P, Lappegård KT, Espevik T, Mollnes TE. The complement system and toll-like receptors as integrated players in the pathophysiology of atherosclerosis. Atherosclerosis 2015; 241:480-94. [PMID: 26086357 DOI: 10.1016/j.atherosclerosis.2015.05.038] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 05/08/2015] [Accepted: 05/29/2015] [Indexed: 02/08/2023]
Abstract
Despite recent medical advances, atherosclerosis is a global burden accounting for numerous deaths and hospital admissions. Immune-mediated inflammation is a major component of the atherosclerotic process, but earlier research focus on adaptive immunity has gradually switched towards the role of innate immunity. The complement system and toll-like receptors (TLRs), and the crosstalk between them, may be of particular interest both with respect to pathogenesis and as therapeutic targets in atherosclerosis. Animal studies indicate that inhibition of C3a and C5a reduces atherosclerosis. In humans modified LDL-cholesterol activate complement and TLRs leading to downstream inflammation, and histopathological studies indicate that the innate immune system is present in atherosclerotic lesions. Moreover, clinical studies have demonstrated that both complement and TLRs are upregulated in atherosclerotic diseases, although interventional trials have this far been disappointing. However, based on recent research showing an intimate interplay between complement and TLRs we propose a model in which combined inhibition of both complement and TLRs may represent a potent anti-inflammatory therapeutic approach to reduce atherosclerosis.
Collapse
Affiliation(s)
- Anders Hovland
- Coronary Care Unit, Division of Internal Medicine, Nordland Hospital, 8092 Bodø, Norway; Institute of Clinical Medicine, University of Tromsø, 9019 Tromsø, Norway.
| | - Lena Jonasson
- Department of Medical and Health Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631 Rigshospitalet, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Arne Yndestad
- Research Institute of Internal Medicine and Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0318 Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine and Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0318 Oslo, Norway
| | - Knut T Lappegård
- Coronary Care Unit, Division of Internal Medicine, Nordland Hospital, 8092 Bodø, Norway; Institute of Clinical Medicine, University of Tromsø, 9019 Tromsø, Norway
| | - Terje Espevik
- Norwegian University of Science and Technology, Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, 7491 Trondheim, Norway
| | - Tom E Mollnes
- Institute of Clinical Medicine, University of Tromsø, 9019 Tromsø, Norway; K.G. Jebsen Inflammation Research Centre, University of Oslo, 0318 Oslo, Norway; Norwegian University of Science and Technology, Centre of Molecular Inflammation Research, and Department of Cancer Research and Molecular Medicine, 7491 Trondheim, Norway; Research Laboratory, Nordland Hospital, 8092 Bodø, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, 0372 Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9019 Tromsø, Norway
| |
Collapse
|
26
|
Merle NS, Noe R, Halbwachs-Mecarelli L, Fremeaux-Bacchi V, Roumenina LT. Complement System Part II: Role in Immunity. Front Immunol 2015; 6:257. [PMID: 26074922 PMCID: PMC4443744 DOI: 10.3389/fimmu.2015.00257] [Citation(s) in RCA: 686] [Impact Index Per Article: 68.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/09/2015] [Indexed: 12/14/2022] Open
Abstract
The complement system has been considered for a long time as a simple lytic cascade, aimed to kill bacteria infecting the host organism. Nowadays, this vision has changed and it is well accepted that complement is a complex innate immune surveillance system, playing a key role in host homeostasis, inflammation, and in the defense against pathogens. This review discusses recent advances in the understanding of the role of complement in physiology and pathology. It starts with a description of complement contribution to the normal physiology (homeostasis) of a healthy organism, including the silent clearance of apoptotic cells and maintenance of cell survival. In pathology, complement can be a friend or a foe. It acts as a friend in the defense against pathogens, by inducing opsonization and a direct killing by C5b–9 membrane attack complex and by triggering inflammatory responses with the anaphylatoxins C3a and C5a. Opsonization plays also a major role in the mounting of an adaptive immune response, involving antigen presenting cells, T-, and B-lymphocytes. Nevertheless, it can be also an enemy, when pathogens hijack complement regulators to protect themselves from the immune system. Inadequate complement activation becomes a disease cause, as in atypical hemolytic uremic syndrome, C3 glomerulopathies, and systemic lupus erythematosus. Age-related macular degeneration and cancer will be described as examples showing that complement contributes to a large variety of conditions, far exceeding the classical examples of diseases associated with complement deficiencies. Finally, we discuss complement as a therapeutic target.
Collapse
Affiliation(s)
- Nicolas S Merle
- UMRS 1138, Centre de Recherche des Cordeliers, INSERM , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, UPMC Université Paris 06 , Paris , France
| | - Remi Noe
- UMRS 1138, Centre de Recherche des Cordeliers, INSERM , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, UPMC Université Paris 06 , Paris , France ; Ecole Pratique des Hautes Études (EPHE) , Paris , France
| | - Lise Halbwachs-Mecarelli
- UMRS 1138, Centre de Recherche des Cordeliers, INSERM , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, UPMC Université Paris 06 , Paris , France
| | - Veronique Fremeaux-Bacchi
- UMRS 1138, Centre de Recherche des Cordeliers, INSERM , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, UPMC Université Paris 06 , Paris , France ; Service d'Immunologie Biologique, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou , Paris , France
| | - Lubka T Roumenina
- UMRS 1138, Centre de Recherche des Cordeliers, INSERM , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Paris Cité, Université Paris Descartes , Paris , France ; UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Universités, UPMC Université Paris 06 , Paris , France
| |
Collapse
|
27
|
Raiten DJ, Sakr Ashour FA, Ross AC, Meydani SN, Dawson HD, Stephensen CB, Brabin BJ, Suchdev PS, van Ommen B. Inflammation and Nutritional Science for Programs/Policies and Interpretation of Research Evidence (INSPIRE). J Nutr 2015; 145:1039S-1108S. [PMID: 25833893 PMCID: PMC4448820 DOI: 10.3945/jn.114.194571] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/08/2014] [Accepted: 10/29/2014] [Indexed: 02/06/2023] Open
Abstract
An increasing recognition has emerged of the complexities of the global health agenda—specifically, the collision of infections and noncommunicable diseases and the dual burden of over- and undernutrition. Of particular practical concern are both 1) the need for a better understanding of the bidirectional relations between nutritional status and the development and function of the immune and inflammatory response and 2) the specific impact of the inflammatory response on the selection, use, and interpretation of nutrient biomarkers. The goal of the Inflammation and Nutritional Science for Programs/Policies and Interpretation of Research Evidence (INSPIRE) is to provide guidance for those users represented by the global food and nutrition enterprise. These include researchers (bench and clinical), clinicians providing care/treatment, those developing and evaluating programs/interventions at scale, and those responsible for generating evidence-based policy. The INSPIRE process included convening 5 thematic working groups (WGs) charged with developing summary reports around the following issues: 1) basic overview of the interactions between nutrition, immune function, and the inflammatory response; 2) examination of the evidence regarding the impact of nutrition on immune function and inflammation; 3) evaluation of the impact of inflammation and clinical conditions (acute and chronic) on nutrition; 4) examination of existing and potential new approaches to account for the impact of inflammation on biomarker interpretation and use; and 5) the presentation of new approaches to the study of these relations. Each WG was tasked with synthesizing a summary of the evidence for each of these topics and delineating the remaining gaps in our knowledge. This review consists of a summary of the INSPIRE workshop and the WG deliberations.
Collapse
Affiliation(s)
- Daniel J Raiten
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD;
| | - Fayrouz A Sakr Ashour
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | - A Catharine Ross
- Departments of Nutritional Sciences and Veterinary and Biomedical Science and Center for Molecular Immunology and Infectious Disease, Pennsylvania State University, University Park, PA
| | - Simin N Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Harry D Dawson
- USDA-Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, Beltsville, MD
| | - Charles B Stephensen
- Agricultural Research Service, Western Human Nutrition Research Center, USDA, Davis, CA
| | - Bernard J Brabin
- Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Liverpool, United Kingdom; Global Child Health Group, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Parminder S Suchdev
- Department of Pediatrics and Global Health, Emory University, Atlanta, GA; and
| | | |
Collapse
|
28
|
Török K, Dezső B, Bencsik A, Uzonyi B, Erdei A. Complement receptor type 1 (CR1/CD35) expressed on activated human CD4+ T cells contributes to generation of regulatory T cells. Immunol Lett 2015; 164:117-24. [PMID: 25742728 DOI: 10.1016/j.imlet.2015.02.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 11/25/2022]
Abstract
The role of complement in the regulation of T cell immunity has been highlighted recently by several groups. We were prompted to reinvestigate the role of complement receptor type 1 (CR1, CD35) [corrected] in human T cells based on our earlier data showing that activated human T cells produce C3 (Torok et al. (2012) [48]) and also by results demonstrating that engagement of Membrane Cofactor Protein (MCP, CD46) induces a switch of anti-CD35-activated [corrected] helper T cells into regulatory T cells (Kemper et al. (2003) [17]). We demonstrate here that co-ligation of CD46 and CD35, [corrected] the two C3b-binding structures present on activated CD4+ human T cells significantly enhances CD25 expression, elevates granzyme B production and synergistically augments cell proliferation. The role of CR1 in the development of the Treg phenotype was further confirmed by demonstrating that its engagement enhances IL-10 production and reduces IFNγ release by the activated CD4+ T cells in the presence of excess IL-2. The functional in vivo relevance of our findings was highlighted by the immunohistochemical staining of tonsils, revealing the presence of CD4/CD35 [corrected] double positive lymphocytes mainly in the inter-follicular regions where direct contact between CD4+ T cells and B lymphocytes occurs. Regarding the in vivo relevance of the complement-dependent generation of regulatory T cells in secondary lymphoid organs we propose a scenario shown in the figure. The depicted process involves the sequential binding of locally produced C3 fragments to CD46 and CD35 [corrected] expressed on activated T cells, which - in the presence of excess IL-2 - leads to the development of Treg cells.
Collapse
Affiliation(s)
- Katalin Török
- MTA-ELTE Immunology Research Group, Budapest, Hungary
| | - Balázs Dezső
- Department of Pathology, Medical Center, University of Debrecen, Hungary
| | | | | | - Anna Erdei
- MTA-ELTE Immunology Research Group, Budapest, Hungary; Department of Immunology, Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
29
|
Jongerius I, Schuijt TJ, Mooi FR, Pinelli E. Complement evasion by Bordetella pertussis: implications for improving current vaccines. J Mol Med (Berl) 2015; 93:395-402. [PMID: 25686752 PMCID: PMC4366546 DOI: 10.1007/s00109-015-1259-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 01/19/2015] [Accepted: 01/27/2015] [Indexed: 12/17/2022]
Abstract
Bordetella pertussis causes whooping cough or pertussis, a highly contagious disease of the respiratory tract. Despite high vaccination coverage, reported cases of pertussis are rising worldwide and it has become clear that the current vaccines must be improved. In addition to the well-known protective role of antibodies and T cells during B. pertussis infection, innate immune responses such as the complement system play an essential role in B. pertussis killing. In order to evade this complement activation and colonize the human host, B. pertussis expresses several molecules that inhibit complement activation. Interestingly, one of the known complement evasion proteins, autotransporter Vag8, is highly expressed in the recently emerged B. pertussis isolates. Here, we describe the current knowledge on how B. pertussis evades complement-mediated killing. In addition, we compare this to complement evasion strategies used by other bacterial species. Finally, we discuss the consequences of complement evasion by B. pertussis on adaptive immunity and how identification of the bacterial molecules and the mechanisms involved in complement evasion might help improve pertussis vaccines.
Collapse
Affiliation(s)
- Ilse Jongerius
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
- Present Address: Department of Medical Microbiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Tim J. Schuijt
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
- Present Address: Department of Clinical Chemistry, Hematology and Immunology, Diakonessenhuis, Bosboomstraat 1, 3582 KE Utrecht, The Netherlands
| | - Frits R. Mooi
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| | - Elena Pinelli
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, P.O. Box 1, 3720 BA Bilthoven, The Netherlands
| |
Collapse
|
30
|
Abstract
Galectins are an evolutionarily ancient family of glycan-binding proteins (GBPs) and are found in all animals. Although they were discovered over 30 years ago, ideas about their biological functions continue to evolve. Current evidence indicates that galectins, which are the only known GBPs that occur free in the cytoplasm and extracellularly, are involved in a variety of intracellular and extracellular pathways contributing to homeostasis, cellular turnover, cell adhesion, and immunity. Here we review evolving insights into galectin biology from a historical perspective and explore current evidence regarding biological roles of galectins.
Collapse
|
31
|
Abstract
Complement is traditionally known to be a system of serum proteins that provide protection against pathogens through direct cell lysis and the mobilization of innate and adaptive immunity. However, recent work indicates that the complement system has additional physiological roles beyond those in host defence. In this Opinion article, we describe the new modes and locations of complement activation that enable it to interact with other cell effector systems, such as growth factor receptors, inflammasomes and metabolic pathways. We propose that the location of complement activation dictates its function.
Collapse
|
32
|
Abstract
Complement is well appreciated to be a potent innate immune defense against microbes and is important in the housekeeping act of removal of apoptotic and effete cells. It is also understood that hyperactivation of complement, or the lack of regulators, may underlie chronic inflammatory diseases. A pipeline of products to intervene in complement activation, some already in clinical use, is being studied in various chronic inflammatory diseases. To date, the role of complement in inflammatory bowel disease has not received a lot of research interest. Novel genetically modified laboratory animals and experiments using antagonists to complement effector molecules have kindled important research observations implicating the complement system in inflammatory bowel disease pathogenesis. We review the evidence base for the role and potential therapeutic manipulation of the complement cascade in inflammatory bowel disease.
Collapse
|
33
|
Novel insights into the regulatory architecture of CD4+ T cells in rheumatoid arthritis. PLoS One 2014; 9:e100690. [PMID: 24959711 PMCID: PMC4069080 DOI: 10.1371/journal.pone.0100690] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/23/2014] [Indexed: 12/21/2022] Open
Abstract
Rheumatoid arthritis (RA) is the most frequent autoimmune chronic inflammatory disease of the joints and it is characterized by the inflammation of the synovial membrane and the subsequent destruction of the joints. In RA, CD4+ T cells are the main drivers of disease initiation and the perpetuation of the damaging inflammatory process. To date, however, the genetic regulatory mechanisms of CD4+ T cells associated with RA etiology are poorly understood. The genome-wide analysis of expression quantitative trait loci (eQTL) in disease-relevant cell types is a recent genomic integration approach that is providing significant insights into the genetic regulatory mechanisms of many human pathologies. The objective of the present study was to analyze, for the first time, the genome-wide genetic regulatory mechanisms associated with the gene expression of CD4+ T cells in RA. Whole genome gene expression profiling of CD4+ T cells and the genome-wide genotyping (598,258 SNPs) of 29 RA patients with an active disease were performed. In order to avoid the excessive burden of multiple testing associated with genome-wide trans-eQTL analysis, we developed and implemented a novel systems genetics approach. Finally, we compared the genomic regulation pattern of CD4+ T cells in RA with the genomic regulation observed in reference lymphoblastoid cell lines (LCLs). We identified a genome-wide significant cis-eQTL associated with the expression of FAM66C gene (P = 6.51e−9). Using our new systems genetics approach we identified six statistically significant trans-eQTLs associated with the expression of KIAA0101 (P<7.4e−8) and BIRC5 (P = 5.35e−8) genes. Finally, comparing the genomic regulation profiles between RA CD4+ T cells and control LCLs we found 20 genes showing differential regulatory patterns between both cell types. The present genome-wide eQTL analysis has identified new genetic regulatory elements that are key to the activity of CD4+ T cells in RA.
Collapse
|
34
|
CD8+DR+ T-Cells and C3 Complement Serum Concentration as Potential Biomarkers in Thrombotic Antiphospholipid Syndrome. Autoimmune Dis 2014; 2014:868652. [PMID: 24982803 PMCID: PMC4058841 DOI: 10.1155/2014/868652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 05/19/2014] [Indexed: 01/08/2023] Open
Abstract
Purpose. To assess complement factors and T lymphocyte activation subset abnormalities in patients with thrombotic antiphospholipid syndrome (APS) as potential biomarkers for development of clinical complications. Methods. We assessed C3, C4, factor B concentrations (nephelometry), complement haemolytic functional activity (CH100, radial immune diffusion), and the activation status of CD4+ and CD8+ T-cells (three-colour flow cytometry) in patients with thrombotic APS. Antiphospholipid (aPL) positive patients without APS-related clinical criteria, systemic lupus erythematosus (SLE) patients, and healthy individuals were evaluated as controls. A clinical followup was performed to assess the potential relationship between the immunological parameters and development of APS-related complications. Results. Lower concentrations of C3 and higher levels of CD8+DR+ cells were risk factors for development of APS-related complications during followup, including rethrombosis and neuropsychiatric symptoms. Patients with diagnosed thrombotic APS had significantly lower levels of C3, C4, and CH100 as well as higher percentages of activated CD4+DR+ and of CD8+DR+ T-cells than healthy controls but similar to that observed in autoimmune disease controls. Conclusion. Lower C3 and C4 complement levels and higher percentages of CD8+DR+ T-cells were observed in thrombotic APS patients. The potential role of these abnormalities as biomarkers of clinical outcome warrants further evaluation in a multicenter study.
Collapse
|
35
|
Xiao F, Ma L, Zhao M, Huang G, Mirenda V, Dorling A, Lechler R, Lombardi G. Ex vivo expanded human regulatory T cells delay islet allograft rejection via inhibiting islet-derived monocyte chemoattractant protein-1 production in CD34+ stem cells-reconstituted NOD-scid IL2rγnull mice. PLoS One 2014; 9:e90387. [PMID: 24594640 PMCID: PMC3940883 DOI: 10.1371/journal.pone.0090387] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 01/28/2014] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by immune-mediated destruction of insulin-secreting β cells of the pancreas. Near complete dependence on exogenous insulin makes T1DM very difficult to control, with the result that patients are exposed to high blood glucose and risk of diabetic complications and/or intermittent low blood glucose that can cause unconsciousness, fits and even death. Allograft transplantation of pancreatic islets restores normoglycemia with a low risk of surgical complications. However, although successful immediately after transplantation, islets are progressively lost, with most of the patients requiring exogenous insulin within 2 years post-transplant. Therefore, there is an urgent requirement for the development of new strategies to prevent islet rejection. In this study, we explored the importance of human regulatory T cells in the control of islets allograft rejection. We developed a pre-clinical model of human islet transplantation by reconstituting NOD-scid IL2rγnull mice with cord blood-derived human CD34+ stem cells and demonstrated that although the engrafted human immune system mediated the rejection of human islets, their survival was significantly prolonged following adoptive transfer of ex vivo expanded human Tregs. Mechanistically, Tregs inhibited the infiltration of innate immune cells and CD4+ T cells into the graft by down-regulating the islet graft-derived monocyte chemoattractant protein-1. Our findings might contribute to the development of clinical strategies for Treg therapy to control human islet rejection. We also show for the first time that CD34+ cells-reconstituted NOD-scid IL2rγnull mouse model could be beneficial for investigating human innate immunity in vivo.
Collapse
Affiliation(s)
- Fang Xiao
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Liang Ma
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Min Zhao
- Department of Diabetes & Endocrinology, King's College London, London, United Kingdom
| | - Guocai Huang
- Department of Diabetes & Endocrinology, King's College London, London, United Kingdom
| | - Vincenzo Mirenda
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Anthony Dorling
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Robert Lechler
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
| | - Giovanna Lombardi
- Medical Research Council (MRC) for Transplantation, King's College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
The role of decay accelerating factor in environmentally induced and idiopathic systemic autoimmune disease. Autoimmune Dis 2014; 2014:452853. [PMID: 24592327 PMCID: PMC3921935 DOI: 10.1155/2014/452853] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/19/2013] [Indexed: 01/05/2023] Open
Abstract
Decay accelerating factor (DAF) plays a complex role in the immune system through complement-dependent and -independent regulation of innate and adaptive immunity. Over the past five years there has been accumulating evidence for a significant role of DAF in negatively regulating adaptive T-cell responses and autoimmunity in both humans and experimental models. This review discusses the relationship between DAF and the complement system and highlights major advances in our understanding of the biology of DAF in human disease, particularly systemic lupus erythematosus. The role of DAF in regulation of idiopathic and environmentally induced systemic autoimmunity is discussed including studies showing that reduction or absence of DAF is associated with autoimmunity. In contrast, DAF-mediated T cell activation leads to cytokine expression consistent with T regulatory cells. This is supported by studies showing that interaction between DAF and its molecular partner, CD97, modifies expression of autoimmunity promoting cytokines. These observations are used to develop a hypothetical model to explain how DAF expression may impact T cell differentiation via interaction with CD97 leading to T regulatory cells, increased production of IL-10, and immune tolerance.
Collapse
|
37
|
Liszewski MK, Kolev M, Le Friec G, Leung M, Bertram PG, Fara AF, Subias M, Pickering MC, Drouet C, Meri S, Arstila TP, Pekkarinen PT, Ma M, Cope A, Reinheckel T, Rodriguez de Cordoba S, Afzali B, Atkinson JP, Kemper C. Intracellular complement activation sustains T cell homeostasis and mediates effector differentiation. Immunity 2013; 39:1143-57. [PMID: 24315997 PMCID: PMC3865363 DOI: 10.1016/j.immuni.2013.10.018] [Citation(s) in RCA: 415] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/19/2013] [Indexed: 01/27/2023]
Abstract
Complement is viewed as a critical serum-operative component of innate immunity, with processing of its key component, C3, into activation fragments C3a and C3b confined to the extracellular space. We report here that C3 activation also occurred intracellularly. We found that the T cell-expressed protease cathepsin L (CTSL) processed C3 into biologically active C3a and C3b. Resting T cells contained stores of endosomal and lysosomal C3 and CTSL and substantial amounts of CTSL-generated C3a. While “tonic” intracellular C3a generation was required for homeostatic T cell survival, shuttling of this intracellular C3-activation-system to the cell surface upon T cell stimulation induced autocrine proinflammatory cytokine production. Furthermore, T cells from patients with autoimmune arthritis demonstrated hyperactive intracellular complement activation and interferon-γ production and CTSL inhibition corrected this deregulated phenotype. Importantly, intracellular C3a was observed in all examined cell populations, suggesting that intracellular complement activation might be of broad physiological significance. Complement C3 is activated intracellularly in human T cells by cathepsin L Intracellular C3 activation mediates cell survival and Th1 induction Increased intracellular C3 activation underlies T effector dysregulation in arthritis Patients with serum C3-deficiency retain intracellular C3a generation
Collapse
Affiliation(s)
- M Kathryn Liszewski
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Martin Kolev
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Gaelle Le Friec
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Marilyn Leung
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Paula G Bertram
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Antonella F Fara
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Marta Subias
- Departamento de Immunología, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28006, Spain
| | - Matthew C Pickering
- Centre for Complement and Inflammation Research, Imperial College, London SW7 2AZ, UK
| | - Christian Drouet
- Université Joseph Fourier, GREPI/AGIM CNRS FRE3405, Grenoble, F-38041, France
| | - Seppo Meri
- Haartman Institute and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, FI-00014, Finland
| | - T Petteri Arstila
- Haartman Institute and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, FI-00014, Finland
| | - Pirkka T Pekkarinen
- Haartman Institute and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, FI-00014, Finland
| | - Margaret Ma
- Biomedical Research Centre, King's Health Partners, Guy's Hospital, London SE1 9RT, UK; Academic Department of Rheumatology, King's College London, London SE1 9RT, UK
| | - Andrew Cope
- Biomedical Research Centre, King's Health Partners, Guy's Hospital, London SE1 9RT, UK; Academic Department of Rheumatology, King's College London, London SE1 9RT, UK
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, and BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, D-79104, Germany
| | - Santiago Rodriguez de Cordoba
- Departamento de Immunología, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28006, Spain
| | - Behdad Afzali
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK; Biomedical Research Centre, King's Health Partners, Guy's Hospital, London SE1 9RT, UK
| | - John P Atkinson
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Claudia Kemper
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK.
| |
Collapse
|
38
|
Yamamoto H, Fara AF, Dasgupta P, Kemper C. CD46: the 'multitasker' of complement proteins. Int J Biochem Cell Biol 2013; 45:2808-20. [PMID: 24120647 DOI: 10.1016/j.biocel.2013.09.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 09/23/2013] [Accepted: 09/30/2013] [Indexed: 12/12/2022]
Abstract
Complement is undeniably quintessential for innate immunity by detecting and eliminating infectious microorganisms. Recent work, however, highlights an equally profound impact of complement on the induction and regulation of a wide range of immune cells. In particular, the complement regulator CD46 emerges as a key sensor of immune activation and a vital modulator of adaptive immunity. In this review, we summarize the current knowledge of CD46-mediated signalling events and their functional consequences on immune-competent cells with a specific focus on those in CD4(+) T cells. We will also discuss the promises and challenges that potential therapeutic modulation of CD46 may hold and pose.
Collapse
Affiliation(s)
- Hidekazu Yamamoto
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London SE1 9RT, UK; The Urology Centre, Guy's and St. Thomas' NHS Foundations Trust, London SE1 9RT, UK
| | | | | | | |
Collapse
|
39
|
C3a modulates IL-1β secretion in human monocytes by regulating ATP efflux and subsequent NLRP3 inflammasome activation. Blood 2013; 122:3473-81. [PMID: 23878142 DOI: 10.1182/blood-2013-05-502229] [Citation(s) in RCA: 244] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Interleukin-1β (IL-1β) is a proinflammatory cytokine and a therapeutic target in several chronic autoimmune states. Monocytes and macrophages are the major sources of IL-1β. IL-1β production by these cells requires Toll-like receptor (TLR) and adenosine triphosphate (ATP)-mediated P2X purinoceptor 7 (P2X7) signals, which together activate the inflammasome. However, how TLR signals and ATP availability are regulated during monocyte activation is unclear and the involvement of another danger signal system has been proposed. Here, we demonstrate that both lipopolysaccharide (LPS) and the anaphylatoxin C3a are needed for IL-1β production in human macrophages and dendritic cells, while in monocytes, C3a enhanced the secretion of LPS-induced IL-1β. C3a and LPS-stimulated monocytes increased T helper 17 (Th17) cell induction in vitro, and human rejecting, but not nonrejecting, kidney transplant biopsies were characterized by local generation of C3a and monocyte and Th17 cell infiltration. Mechanistically, C3a drives IL-1β production in monocytes by controlling the release of intracellular ATP into the extracellular space via regulation of as-yet unidentified ATP-releasing channels in an extracellular signal-regulated kinase 1/2-dependent fashion. These data define a novel function for complement in inflammasome activation in monocytes and suggest that C3aR-mediated signaling is a vital component of the IL-1β-Th17 axis.
Collapse
|