1
|
Stark K, Kilani B, Stockhausen S, Busse J, Schubert I, Tran TD, Gaertner F, Leunig A, Pekayvaz K, Nicolai L, Fumagalli V, Stermann J, Stephan F, David C, Müller MB, Heyman B, Lux A, da Palma Guerreiro A, Frenzel LP, Schmidt CQ, Dopler A, Moser M, Chandraratne S, von Brühl ML, Lorenz M, Korff T, Rudelius M, Popp O, Kirchner M, Mertins P, Nimmerjahn F, Iannacone M, Sperandio M, Engelmann B, Verschoor A, Massberg S. Antibodies and complement are key drivers of thrombosis. Immunity 2024; 57:2140-2156.e10. [PMID: 39226900 DOI: 10.1016/j.immuni.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/17/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Venous thromboembolism (VTE) is a common, deadly disease with an increasing incidence despite preventive efforts. Clinical observations have associated elevated antibody concentrations or antibody-based therapies with thrombotic events. However, how antibodies contribute to thrombosis is unknown. Here, we show that reduced blood flow enabled immunoglobulin M (IgM) to bind to FcμR and the polymeric immunoglobulin receptor (pIgR), initiating endothelial activation and platelet recruitment. Subsequently, the procoagulant surface of activated platelets accommodated antigen- and FcγR-independent IgG deposition. This leads to classical complement activation, setting in motion a prothrombotic vicious circle. Key elements of this mechanism were present in humans in the setting of venous stasis as well as in the dysregulated immunothrombosis of COVID-19. This antibody-driven thrombosis can be prevented by pharmacologically targeting complement. Hence, our results uncover antibodies as previously unrecognized central regulators of thrombosis. These findings carry relevance for therapeutic application of antibodies and open innovative avenues to target thrombosis without compromising hemostasis.
Collapse
Affiliation(s)
- Konstantin Stark
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.
| | - Badr Kilani
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sven Stockhausen
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Johanna Busse
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Irene Schubert
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thuy-Duong Tran
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Florian Gaertner
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany; Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Alexander Leunig
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Valeria Fumagalli
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Julia Stermann
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Felix Stephan
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Christian David
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine, Biomedical Center (BMC) LMU Munich, Munich, Germany
| | - Martin B Müller
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany; Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anja Lux
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Alexandra da Palma Guerreiro
- Department I of Internal Medicine, University Hospital Cologne, Cologne 50937, Germany; Center of Integrated Oncology ABCD, University Hospital of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50937, Germany
| | - Lukas P Frenzel
- Department I of Internal Medicine, University Hospital Cologne, Cologne 50937, Germany; Center of Integrated Oncology ABCD, University Hospital of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50937, Germany
| | - Christoph Q Schmidt
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
| | - Arthur Dopler
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
| | - Markus Moser
- Department of Molecular Medicine, Max-Planck-Institute of Biochemistry, Martinsried, Germany; Institute of Experimental Hematology, TranslaTUM, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Sue Chandraratne
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Marie-Luise von Brühl
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Lorenz
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thomas Korff
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Martina Rudelius
- Institute of Pathology, Ludwig-Maximilian University, Munich, Germany
| | - Oliver Popp
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Matteo Iannacone
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Markus Sperandio
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine, Biomedical Center (BMC) LMU Munich, Munich, Germany
| | - Bernd Engelmann
- Institut für Laboratoriumsmedizin, University Hospital, LMU Munich, Munich, Germany
| | - Admar Verschoor
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany; Department of Otorhinolaryngology, Technische Universität München and Klinikum Rechts der Isar, Munich, Germany.
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
2
|
Tang Y, Luo Y. Identification of a novel mutation in complement receptor 2 in Chinese familial systemic lupus erythematosus. Arch Rheumatol 2022; 37:566-573. [PMID: 36879571 PMCID: PMC9985375 DOI: 10.46497/archrheumatol.2022.9167] [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: 08/29/2021] [Accepted: 12/23/2021] [Indexed: 03/08/2023] Open
Abstract
Objectives This study aims to analyze the relationship between complement receptor 2 (CR2) gene mutation and the clinical phenotype in Chinese familial systemic lupus erythematosus (SLE). Patients and methods A total of one Chinese familial SLE patients (median age: 30.25 years; range, 22 to 49 years) were included between January 2017 and December 2018. The clinical features and diagnoses of familial SLE patients were analyzed using whole-exome sequencing (WES) of genomic deoxyribonucleic acid (DNA) samples. Sanger sequencing was used to verify candidate mutations detected in the examined family. Results The mother and her three daughters were diagnosed with SLE. The clinical characteristics showed that the patient and her mother were diagnosed with lupus nephritis. The eldest daughter had decreased renal function and lower serum albumin levels. Immunological index analysis showed that all four patients were positive for anti-SSA and antinuclear antibody (ANA), but that only the second daughter was positive for anti-double-stranded DNA (dsDNA). Complement 3 (C3) was significantly decreased in all patients, while evaluation of the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) showed that the second and third daughters had mild active SLE. The mother and eldest daughter were treated with prednisolone combined with cyclophosphamide, while the other two daughters were treated with prednisolone alone. The WES and Sanger sequencing analyses revealed an unreported missense T>C mutation c.2804 in the 15th exon of the CR gene in all four patients. Conclusion We identified a novel c.2804 (exon 15) T>C mutation in the CR gene of Chinese familial SLE. This mutation was previously reported, suggesting that the CR gene c.2804 (exon 15) T>C mutation is the probable cause of SLE in this family.
Collapse
Affiliation(s)
- Yuewu Tang
- Department of Nephrology, Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yi Luo
- Department of Blood Transfusion, Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
3
|
Qi R, Qin W. Role of Complement System in Kidney Transplantation: Stepping From Animal Models to Clinical Application. Front Immunol 2022; 13:811696. [PMID: 35281019 PMCID: PMC8913494 DOI: 10.3389/fimmu.2022.811696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/31/2022] [Indexed: 12/23/2022] Open
Abstract
Kidney transplantation is a life-saving strategy for patients with end-stage renal diseases. Despite the advances in surgical techniques and immunosuppressive agents, the long-term graft survival remains a challenge. Growing evidence has shown that the complement system, part of the innate immune response, is involved in kidney transplantation. Novel insights highlighted the role of the locally produced and intracellular complement components in the development of inflammation and the alloreactive response in the kidney allograft. In the current review, we provide the updated understanding of the complement system in kidney transplantation. We will discuss the involvement of the different complement components in kidney ischemia-reperfusion injury, delayed graft function, allograft rejection, and chronic allograft injury. We will also introduce the existing and upcoming attempts to improve allograft outcomes in animal models and in the clinical setting by targeting the complement system.
Collapse
Affiliation(s)
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
4
|
Yarmoska SK, Alawieh AM, Tomlinson S, Hoang KB. Modulation of the Complement System by Neoplastic Disease of the Central Nervous System. Front Immunol 2021; 12:689435. [PMID: 34671342 PMCID: PMC8521155 DOI: 10.3389/fimmu.2021.689435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/10/2021] [Indexed: 12/28/2022] Open
Abstract
The complement system is a highly conserved component of innate immunity that is involved in recognizing and responding to pathogens. The system serves as a bridge between innate and adaptive immunity, and modulation of the complement system can affect the entire host immune response to a foreign insult. Neoplastic diseases have been shown to engage the complement system in order to evade the immune system, gain a selective growth advantage, and co-opt the surrounding environment for tumor proliferation. Historically, the central nervous system has been considered to be an immune-privileged environment, but it is now clear that there are active roles for both innate and adaptive immunity within the central nervous system. Much of the research on the role of immunological modulation of neoplastic disease within the central nervous system has focused on adaptive immunity, even though innate immunity still plays a critical role in the natural history of central nervous system neoplasms. Here, we review the modulation of the complement system by a variety of neoplastic diseases of the central nervous system. We also discuss gaps in the current body of knowledge and comment on future directions for investigation.
Collapse
Affiliation(s)
- Steven K. Yarmoska
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Ali M. Alawieh
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Kimberly B. Hoang
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
5
|
Halting targeted and collateral damage to red blood cells by the complement system. Semin Immunopathol 2021; 43:799-816. [PMID: 34191092 PMCID: PMC8243056 DOI: 10.1007/s00281-021-00859-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/18/2021] [Indexed: 12/12/2022]
Abstract
The complement system is an important defense mechanism against pathogens; however, in certain pathologies, the system also attacks human cells, such as red blood cells (RBCs). In paroxysmal nocturnal hemoglobinuria (PNH), RBCs lack certain complement regulators which sensitize them to complement-mediated lysis, while in autoimmune hemolytic anemia (AIHA), antibodies against RBCs may initiate complement-mediated hemolysis. In recent years, complement inhibition has improved treatment prospects for these patients, with eculizumab now the standard of care for PNH patients. Current complement inhibitors are however not sufficient for all patients, and they come with high costs, patient burden, and increased infection risk. This review gives an overview of the underlying pathophysiology of complement-mediated hemolysis in PNH and AIHA, the role of therapeutic complement inhibition nowadays, and the high number of complement inhibitors currently under investigation, as for almost every complement protein, an inhibitor is being developed. The focus lies with novel therapeutics that inhibit complement activity specifically in the pathway that causes pathology or those that reduce costs or patient burden through novel administration routes.
Collapse
|
6
|
Annamalai B, Parsons N, Nicholson C, Joseph K, Coughlin B, Yang X, Jones BW, Tomlinson S, Rohrer B. Natural immunoglobulin M-based delivery of a complement alternative pathway inhibitor in mouse models of retinal degeneration. Exp Eye Res 2021; 207:108583. [PMID: 33878326 PMCID: PMC8504679 DOI: 10.1016/j.exer.2021.108583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 11/23/2022]
Abstract
PURPOSE Age-related macular degeneration is a slowly progressing disease. Studies have tied disease risk to an overactive complement system. We have previously demonstrated that pathology in two mouse models, the choroidal neovascularization (CNV) model and the smoke-induced ocular pathology (SIOP) model, can be reduced by specifically inhibiting the alternative complement pathway (AP). Here we report on the development of a novel injury-site targeted inhibitor of the alternative pathway, and its characterization in models of retinal degeneration. METHODS Expression of the danger associated molecular pattern, a modified annexin IV, in injured ARPE-19 cells was confirmed by immunohistochemistry and complementation assays using B4 IgM mAb. Subsequently, a construct was prepared consisting of B4 single chain antibody (scFv) linked to a fragment of the alternative pathway inhibitor, fH (B4-scFv-fH). ARPE-19 cells stably expressing B4-scFv-fH were microencapsulated and administered intravitreally or subcutaneously into C57BL/6 J mice, followed by CNV induction or smoke exposure. Progression of CNV was analyzed using optical coherence tomography, and SIOP using structure-function analyses. B4-scFv-fH targeting and AP specificity was assessed by Western blot and binding experiments. RESULTS B4-scFv-fH was secreted from encapsulated RPE and inhibited complement in RPE monolayers. B4-scFv-fH capsules reduced CNV and SIOP, and western blotting for breakdown products of C3α, IgM and IgG confirmed a reduction in complement activation and antibody binding in RPE/choroid. CONCLUSIONS Data supports a role for natural antibodies and neoepitope expression in ocular disease, and describes a novel strategy to target AP-specific complement inhibition to diseased tissue in the eye. PRECIS AMD risk is tied to an overactive complement system, and ocular injury is reduced by alternative pathway (AP) inhibition in experimental models. We developed a novel inhibitor of the AP that targets an injury-specific danger associated molecular pattern, and characterized it in disease models.
Collapse
Affiliation(s)
| | - Nathaniel Parsons
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
| | - Crystal Nicholson
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
| | - Kusumam Joseph
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
| | - Beth Coughlin
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA
| | - Xiaofeng Yang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Bryan W Jones
- Department of Ophthalmology, University of Utah, Salt Lake City, UT, USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA; Ralph H. Johnson VA Medical Center, Division of Research, Charleston, SC, USA
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA; Ralph H. Johnson VA Medical Center, Division of Research, Charleston, SC, USA; Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
7
|
Regulation of the complement system and immunological tolerance in pregnancy. Semin Immunol 2019; 45:101337. [PMID: 31757607 DOI: 10.1016/j.smim.2019.101337] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/24/2019] [Indexed: 12/18/2022]
Abstract
Preeclampsia is a serious vascular complication of the human pregnancy, whose etiology is still poorly understood. In preeclampsia, exacerbated apoptosis and fragmentation of the placental tissue occurs due to developmental qualities of the placental trophoblast cells and/or mechanical and oxidative distress to the syncytiotrophoblast, which lines the placental villi. Dysregulation of the complement system is recognized as one of the mechanisms of the disease pathology. Complement has the ability to promote inflammation and facilitate phagocytosis of placenta-derived particles and apoptotic cells by macrophages. In preeclampsia, an overload of placental cell damage or dysregulated complement system may lead to insufficient clearance of apoptotic particles and placenta-derived debris. Excess placental damage may lead to sequestration of microparticles, such as placental vesicles, to capillaries in the glomeruli of the kidney and other vulnerable tissues. This phenomenon could contribute to the manifestations of typical diagnostic symptoms of preeclampsia: proteinuria and new-onset hypertension. In this review we propose that the complement system may serve as a regulator of the complex tolerance and clearance processes that are fundamental in healthy pregnancy. It is therefore recommended that further research be conducted to elucidate the interactions between components of the complement system and immune responses in the context of complicated and healthy pregnancy.
Collapse
|
8
|
Ma Y, Liu Y, Zhang Z, Yang GY. Significance of Complement System in Ischemic Stroke: A Comprehensive Review. Aging Dis 2019; 10:429-462. [PMID: 31011487 PMCID: PMC6457046 DOI: 10.14336/ad.2019.0119] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/19/2019] [Indexed: 12/14/2022] Open
Abstract
The complement system is an essential part of innate immunity, typically conferring protection via eliminating pathogens and accumulating debris. However, the defensive function of the complement system can exacerbate immune, inflammatory, and degenerative responses in various pathological conditions. Cumulative evidence indicates that the complement system plays a critical role in the pathogenesis of ischemic brain injury, as the depletion of certain complement components or the inhibition of complement activation could reduce ischemic brain injury. Although multiple candidates modulating or inhibiting complement activation show massive potential for the treatment of ischemic stroke, the clinical availability of complement inhibitors remains limited. The complement system is also involved in neural plasticity and neurogenesis during cerebral ischemia. Thus, unexpected side effects could be induced if the systemic complement system is inhibited. In this review, we highlighted the recent concepts and discoveries of the roles of different kinds of complement components, such as C3a, C5a, and their receptors, in both normal brain physiology and the pathophysiology of brain ischemia. In addition, we comprehensively reviewed the current development of complement-targeted therapy for ischemic stroke and discussed the challenges of bringing these therapies into the clinic. The design of future experiments was also discussed to better characterize the role of complement in both tissue injury and recovery after cerebral ischemia. More studies are needed to elucidate the molecular and cellular mechanisms of how complement components exert their functions in different stages of ischemic stroke to optimize the intervention of targeting the complement system.
Collapse
Affiliation(s)
- Yuanyuan Ma
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqun Liu
- 3Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhijun Zhang
- 2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Hammad A, Westacott L, Zaben M. The role of the complement system in traumatic brain injury: a review. J Neuroinflammation 2018; 15:24. [PMID: 29357880 PMCID: PMC5778697 DOI: 10.1186/s12974-018-1066-z] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/15/2018] [Indexed: 02/08/2023] Open
Abstract
Traumatic brain injury (TBI) is an important cause of disability and mortality in the western world. While the initial injury sustained results in damage, it is the subsequent secondary cascade that is thought to be the significant determinant of subsequent outcomes. The changes associated with the secondary injury do not become irreversible until some time after the start of the cascade. This may present a window of opportunity for therapeutic interventions aiming to improve outcomes subsequent to TBI. A prominent contributor to the secondary injury is a multifaceted inflammatory reaction. The complement system plays a notable role in this inflammatory reaction; however, it has often been overlooked in the context of TBI secondary injury. The complement system has homeostatic functions in the uninjured central nervous system (CNS), playing a part in neurodevelopment as well as having protective functions in the fully developed CNS, including protection from infection and inflammation. In the context of CNS injury, it can have a number of deleterious effects, evidence for which primarily comes not only from animal models but also, to a lesser extent, from human post-mortem studies. In stark contrast to this, complement may also promote neurogenesis and plasticity subsequent to CNS injury. This review aims to explore the role of the complement system in TBI secondary injury, by examining evidence from both clinical and animal studies. We examine whether specific complement activation pathways play more prominent roles in TBI than others. We also explore the potential role of complement in post-TBI neuroprotection and CNS repair/regeneration. Finally, we highlight the therapeutic potential of targeting the complement system in the context of TBI and point out certain areas on which future research is needed.
Collapse
Affiliation(s)
- Adnan Hammad
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Laura Westacott
- Neuroscience and Mental Health Research Institute (NMHRI), School of Medicine, Cardiff University, Room 4FT 80E, 4th Floor, Heath Park, Cardiff, CF14 4XN UK
| | - Malik Zaben
- Neuroscience and Mental Health Research Institute (NMHRI), School of Medicine, Cardiff University, Room 4FT 80E, 4th Floor, Heath Park, Cardiff, CF14 4XN UK
| |
Collapse
|
10
|
Rich MC, Keene CN, Neher MD, Johnson K, Yu ZX, Ganivet A, Holers VM, Stahel PF. Site-targeted complement inhibition by a complement receptor 2-conjugated inhibitor (mTT30) ameliorates post-injury neuropathology in mouse brains. Neurosci Lett 2016; 617:188-94. [PMID: 26892188 DOI: 10.1016/j.neulet.2016.02.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 02/10/2016] [Accepted: 02/12/2016] [Indexed: 11/26/2022]
Abstract
Intracerebral complement activation after severe traumatic brain injury (TBI) leads to a cascade of neuroinflammatory pathological sequelae that propagate host-mediated secondary brain injury and adverse outcomes. There are currently no specific pharmacological agents on the market to prevent or mitigate the development of secondary cerebral insults after TBI. A novel chimeric CR2-fH compound (mTT30) provides targeted inhibition of the alternative complement pathway at the site of tissue injury. This experimental study was designed to test the neuroprotective effects of mTT30 in a mouse model of closed head injury. The administration of 500 μg mTT30 i.v. at 1 h, 4 h and 24 h after head injury attenuated complement C3 deposition in injured brains, reduced the extent of neuronal cell death, and decreased post-injury microglial activation, compared to vehicle-injected placebo controls. These data imply that site-targeted alternative pathway complement inhibition may represent a new promising therapeutic avenue for the future management of severe TBI.
Collapse
Affiliation(s)
- Megan C Rich
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA
| | - Chesleigh N Keene
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA
| | - Miriam D Neher
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA
| | | | - Zhao-Xue Yu
- Alexion Pharmaceuticals, Cheshire, CT 06410, USA
| | - Antoine Ganivet
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA
| | - V Michael Holers
- Department of Medicine and Department of Immunology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Philip F Stahel
- Department of Orthopaedic Surgery, Denver Health Medical Center and University of Colorado School of Medicine, Denver, CO 80204, USA; Department of Neurosurgery, University of Colorado School of Medicine, Denver, CO 80204, USA.
| |
Collapse
|
11
|
|
12
|
Atkinson C, Qiao F, Yang X, Zhu P, Reaves N, Kulik L, Goddard M, Holers VM, Tomlinson S. Targeting pathogenic postischemic self-recognition by natural IgM to protect against posttransplantation cardiac reperfusion injury. Circulation 2015; 131:1171-80. [PMID: 25825397 DOI: 10.1161/circulationaha.114.010482] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Natural IgM antibodies represent a class of innate pattern recognition receptors that recognize danger-associated molecular patterns expressed on stressed or dying cells. They play important roles in tissue homeostasis by disposing of prenecrotic cells and suppressing inflammation. However, ischemic insult leads to a pathogenic level of IgM binding and complement activation, resulting in inflammation and injury. We investigate the role of self-reactive IgM in the unique setting of transplantation where the donor organ undergoes both cold and warm ischemia and global ischemic insult. METHODS AND RESULTS By transplanting hearts from wild-type donor mice into antibody-deficient mice reconstituted with specific self-reactive IgM monoclonal antibodies, we identified neoepitopes expressed after transplantation and demonstrated a key role for IgM recognition of these epitopes in graft injury. With this information, we developed and characterized a therapeutic strategy that exploited the postischemia recognition system of natural antibodies. On the basis of neoepitope identification, we constructed an anti-annexin IV single-chain antibody (scFv) and an scFv linked to Crry, an inhibitor of C3 activation (scFv-Crry). In an allograft transplantation model in which recipients contain a full natural antibody repertoire, both constructs blocked graft IgM binding and complement activation and significantly reduced graft inflammation and injury. Furthermore, scFv-Crry specifically targeted to the transplanted heart and, unlike complement deficiency, did not affect immunity to infection, an important consideration for immunosuppressed transplant recipients. CONCLUSIONS We identified pathophysiologically important epitopes expressed within the heart after transplantation and described a novel translatable strategy for targeted complement inhibition that has several advantages over currently available approaches.
Collapse
Affiliation(s)
- Carl Atkinson
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Fei Qiao
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Xiaofeng Yang
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Peng Zhu
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Nicholas Reaves
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Liudmila Kulik
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Martin Goddard
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - V Michael Holers
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.)
| | - Stephen Tomlinson
- From Department of Microbiology and Immunology, Medical University of South Carolina, Charleston (CA., F.Q., X.Y., P.Z., N.R., S.T.); Department of Medicine and Immunology, University of Colorado Denver, Aurora (L.K., V.M.H.); Department of Pathology, Papworth Hospital, Cambridgeshire, UK (M.G.); and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC (S.T.).
| |
Collapse
|
13
|
Zhang S, Cui P. Complement system in zebrafish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 46:3-10. [PMID: 24462834 DOI: 10.1016/j.dci.2014.01.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/08/2014] [Accepted: 01/08/2014] [Indexed: 06/03/2023]
Abstract
Zebrafish is recently emerging as a model species for the study of immunology and human diseases. Complement system is the humoral backbone of the innate immune defense, and our knowledge as such in zebrafish has dramatically increased in the recent years. This review summarizes the current research progress of zebrafish complement system. The global searching for complement components in genome database, together with published data, has unveiled the existence of all the orthologues of mammalian complement components identified thus far, including the complement regulatory proteins and complement receptors, in zebrafish. Interestingly, zebrafish complement components also display some distinctive features, such as prominent levels of extrahepatic expression and isotypic diversity of the complement components. Future studies should focus on the following issues that would be of special importance for understanding the physiological role of complement components in zebrafish: conclusive identification of complement genes, especially those with isotypic diversity; analysis and elucidation of function and mechanism of complement components; modulation of innate and adaptive immune response by complement system; and unconventional roles of complement-triggered pathways.
Collapse
Affiliation(s)
- Shicui Zhang
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China.
| | - Pengfei Cui
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, China
| |
Collapse
|
14
|
Abstract
The complement system plays a major role in the autoimmune disease, systemic lupus erythematosus (SLE). However, the role of complement in SLE is complex since it may both prevent and exacerbate the disease. In this review, we explore the latest findings in complement-focused research in SLE. C1q deficiency is the strongest genetic risk factor for SLE, although such deficiency is very rare. Various recently discovered genetic associations include mutations in the complement receptors 2 and 3 as well as complement inhibitors, the latter related to earlier onset of nephritis. Further, autoantibodies are a distinct feature of SLE that are produced as the result of an adaptive immune response and how complement can affect that response is also being reviewed. SLE generates numerous disease manifestations involving contributions from complement such as glomerulonephritis and the increased risk of thrombosis. Furthermore, since most of the complement system is present in plasma, complement is very accessible and may be suitable as biomarker for diagnosis or monitoring of disease activity. This review highlights the many roles of complement for SLE pathogenesis and how research has progressed during recent years.
Collapse
Affiliation(s)
- Jonatan Leffler
- Division of Medical Protein Chemistry, Department of Laboratory Medicine Malmö, Lund University, Malmö, Sweden Division of Cell Biology and Immunology, Telethon Kids Institute, University of Western Australia, Subiaco, Australia
| | - Anders A Bengtsson
- Department of Clinical Sciences, Section of Rheumatology, Lund University, Skåne University Hospital Lund, Lund, Sweden
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Laboratory Medicine Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
15
|
Deficiency of complement receptors CR2/CR1 in Cr2⁻/⁻ mice reduces the extent of secondary brain damage after closed head injury. J Neuroinflammation 2014; 11:95. [PMID: 24885042 PMCID: PMC4050415 DOI: 10.1186/1742-2094-11-95] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 05/12/2014] [Indexed: 12/17/2022] Open
Abstract
Complement activation at the C3 convertase level has been associated with acute neuroinflammation and secondary brain injury after severe head trauma. The present study was designed to test the hypothesis that Cr2-/- mice, which lack the receptors CR2/CD21 and CR1/CD35 for complement C3-derived activation fragments, are protected from adverse sequelae of experimental closed head injury. Adult wild-type mice and Cr2-/- mice on a C57BL/6 genetic background were subjected to focal closed head injury using a standardized weight-drop device. Head-injured Cr2-/- mice showed significantly improved neurological outcomes for up to 72 hours after trauma and a significantly decreased post-injury mortality when compared to wild-type mice. In addition, the Cr2-/- genotype was associated with a decreased extent of neuronal cell death at seven days post-injury. Western blot analysis revealed that complement C3 levels were reduced in the injured brain hemispheres of Cr2-/- mice, whereas plasma C3 levels remained unchanged, compared to wild-type mice. Finally, head-injured Cr2-/- had an attenuated extent of post-injury C3 tissue deposition, decreased astrocytosis and microglial activation, and attenuated immunoglobulin M deposition in injured brains compared to wild-type mice. Targeting of these receptors for complement C3 fragments (CR2/CR1) may represent a promising future approach for therapeutic immunomodulation after traumatic brain injury.
Collapse
|
16
|
Cai Y, Xu H, Yan J, Zhang L, Lu Y. Molecular targets and mechanism of action of dexmedetomidine in treatment of ischemia/reperfusion injury. Mol Med Rep 2014; 9:1542-50. [PMID: 24627001 DOI: 10.3892/mmr.2014.2034] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 02/17/2014] [Indexed: 01/13/2023] Open
Abstract
Dexmedetomidine (DEX), a highly specific α2-adrenergic agonist, which exhibits anaesthetic-sparing, analgesia and sympatholytic properties. DEX modulates gene expression, channel activation, transmitter release, inflammatory processes and apoptotic and necrotic cell death. It has also been demonstrated to have protective effects in a variety of animal models of ischemia/reperfusion (I/R) injury, including the intestine, myocardial, renal, lung, cerebral and liver. The broad spectrum of biological activities associated with DEX continues to expand, and its diverse effects suggest that it may offer a novel therapeutic approach for the treatment of human diseases with I/R involvement.
Collapse
Affiliation(s)
- Ye Cai
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Hui Xu
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Jia Yan
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Lei Zhang
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Yi Lu
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
17
|
Targeting the complement system in systemic lupus erythematosus and other diseases. Clin Immunol 2013; 148:313-21. [DOI: 10.1016/j.clim.2013.02.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 02/22/2013] [Accepted: 02/25/2013] [Indexed: 02/04/2023]
|
18
|
Du Clos TW, Mold C. Complement in host deficiencies and diseases. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
19
|
The increased expression of CD21 on AchR specified B cells in patients with myasthenia gravis. J Neuroimmunol 2012; 256:49-54. [PMID: 23266128 DOI: 10.1016/j.jneuroim.2012.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 11/28/2012] [Accepted: 11/30/2012] [Indexed: 12/15/2022]
Abstract
CD21, a major complement receptor expressed on B cells, is associated with autoimmune disorders. In the present study, we investigated the role of CD21 in pathogenesis of myasthenia gravis (MG) in relationship to anti-acetylcholine receptor (AchR) IgG (anti-AchR IgG) secretion. We detected increased surface expression of CD21 on AchR specified B cells as well as decreased surface expression of CD21 on total B cells in peripheral blood of patients with generalized MG (gMG). In addition, the serum concentrations of soluble secreted CD21 (sCD21) were decreased in patients with gMG. We also found that the level of CD21(+) AchR specified B cells correlated positively with serum anti-AchR IgG level, while the serum concentration of soluble CD21 correlated negatively with serum anti-AchR IgG level. Our data suggests that CD21 might facilitate its function on AchR specified B cell activation, resulting in the secretion of anti-AchR IgG.
Collapse
|
20
|
Brennan FH, Anderson AJ, Taylor SM, Woodruff TM, Ruitenberg MJ. Complement activation in the injured central nervous system: another dual-edged sword? J Neuroinflammation 2012; 9:137. [PMID: 22721265 PMCID: PMC3464784 DOI: 10.1186/1742-2094-9-137] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 06/21/2012] [Indexed: 11/28/2022] Open
Abstract
The complement system, a major component of the innate immune system, is becoming increasingly recognised as a key participant in physiology and disease. The awareness that immunological mediators support various aspects of both normal central nervous system (CNS) function and pathology has led to a renaissance of complement research in neuroscience. Various studies have revealed particularly novel findings on the wide-ranging involvement of complement in neural development, synapse elimination and maturation of neural networks, as well as the progression of pathology in a range of chronic neurodegenerative disorders, and more recently, neurotraumatic events, where rapid disruption of neuronal homeostasis potently triggers complement activation. The purpose of this review is to summarise recent findings on complement activation and acquired brain or spinal cord injury, i.e. ischaemic-reperfusion injury or stroke, traumatic brain injury (TBI) and spinal cord injury (SCI), highlighting the potential for complement-targeted therapeutics to alleviate the devastating consequences of these neurological conditions.
Collapse
Affiliation(s)
- Faith H Brennan
- The University of Queensland, School of Biomedical Sciences, St Lucia, Brisbane, QLD 4072, Australia
| | | | | | | | | |
Collapse
|
21
|
Weckbach S, Neher M, Losacco JT, Bolden AL, Kulik L, Flierl MA, Bell SE, Holers VM, Stahel PF. Challenging the role of adaptive immunity in neurotrauma: Rag1(-/-) mice lacking mature B and T cells do not show neuroprotection after closed head injury. J Neurotrauma 2012; 29:1233-42. [PMID: 22335783 DOI: 10.1089/neu.2011.2169] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The role of adaptive immunity in contributing to post-traumatic neuroinflammation and neuropathology after head injury remains largely unexplored. The present study was designed to investigate the pathophysiological sequelae of closed head injury in Rag1(-/-) mice devoid of mature B and T lymphocytes. C57BL/6 wild-type and Rag1(-/-) mice were subjected to experimental closed head injury, using a standardized weight-drop device. Outcome parameters consisted of neurological scoring, quantification of blood-brain barrier (BBB) function, measurement of inflammatory markers and mediators of apoptosis in serum and brain tissue, and assessment of neuronal cell death, astrogliosis, and tissue destruction. There was no difference between wild-type and Rag1(-/-) mice with regard to injury severity and neurological impairment for up to 7 days after head injury. The extent of BBB dysfunction was in a similar range for both groups. Quantification of complement activation fragments in serum revealed significantly attenuated C3a levels in Rag1(-/-) mice compared to wild-type animals. In contrast, the levels of pro- and anti-inflammatory cytokines and pro-apoptotic and anti-apoptotic mediators remained in a similar range for both groups, and the histological analysis of brain sections did not reveal a difference in reactive astrogliosis, tissue destruction, and neuronal cell death in Rag1(-/-) compared to wild-type mice. These findings suggest that adaptive immunity is not of crucial importance for initiating and sustaining the inflammatory neuropathology after closed head injury. The attenuated extent of post-traumatic complement activation seen in Rag1(-/-) mice implies a cross-talk between innate and adaptive immune responses, which requires further investigation in future studies.
Collapse
Affiliation(s)
- Sebastian Weckbach
- Department of Orthopaedics, Denver Health Medical Center, University of Colorado School of Medicine, Denver, Colorado 80204, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Fischer K, Holt D, Currie B, Kemp D. Scabies: important clinical consequences explained by new molecular studies. ADVANCES IN PARASITOLOGY 2012; 79:339-73. [PMID: 22726646 DOI: 10.1016/b978-0-12-398457-9.00005-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In 2004, we reviewed the status of disease caused by the scabies mite Sarcoptes scabiei at the time and pointed out that very little basic research had ever been done. The reason for this was largely the lack of availability of mites for experimental purposes and, to a degree, a consequent lack of understanding of its importance, resulting in the trivial name 'itch mite'. Scabies is responsible for major morbidity in disadvantaged communities and immunocompromised patients worldwide. In addition to the physical discomfort caused by the disease, scabies infestations facilitate infection by bacterial pathogens such as Streptococcus pyogenes and Staphylococcus aureus via skin lesions, resulting in severe downstream disease such as in a high prevalence of rheumatic fever/heart disease in affected communities. We now have further evidence that in disadvantaged populations living in tropical climates, scabies rather than 'Strep throat' is an important source of S. pyogenes causing rheumatic fever and eventually rheumatic heart disease. In addition, our work has resulted in two fundamental research tools that facilitate much of the current biomedical research efforts on scabies, namely a public database containing ~45,000 scabies mite expressed sequence tags and a porcine in vivo model. Here we will discuss novel and unexpected proteins encountered in the database that appear crucial to mite survival with regard to digestion and evasion of host defence. The mode(s) of action of some of these have been at least partially revealed. Further, newly discovered molecules that may well have a similar role, such as a family of inactivated cysteine proteases, are yet to be investigated. Hence, there are now whole families of potential targets for chemical inhibitors of S. scabiei. These efforts put today's scabies research in a unique position to design and test small molecules that may specifically interfere with mite-derived molecules, such as digestive proteases and mite complement inhibitors. The porcine scabies model will be available to trial in vivo treatment with potential inhibitors. New therapies for scabies may be developed from these studies and may contribute to reduce the spread of scabies and the subsequent prevalence of bacterial skin infections and their devastating sequelae in the community.
Collapse
Affiliation(s)
- Katja Fischer
- Queensland Institute of Medical Research, Herston, Austraria
| | | | | | | |
Collapse
|
23
|
Fleming SD. Naturally Occurring Autoantibodies Mediate Ischemia/Reperfusion-Induced Tissue Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 750:174-85. [DOI: 10.1007/978-1-4614-3461-0_13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
24
|
Nakao M, Tsujikura M, Ichiki S, Vo TK, Somamoto T. The complement system in teleost fish: progress of post-homolog-hunting researches. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:1296-1308. [PMID: 21414344 DOI: 10.1016/j.dci.2011.03.003] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 01/05/2011] [Accepted: 03/06/2011] [Indexed: 05/30/2023]
Abstract
Studies on the complement system of bony fish are now finishing a stage of homologue-hunting identification of the components, unveiling existence of almost all the orthologues of mammalian complement components in teleost. Genomic and transcriptomic data for several teleost species have contributed much for the homologue-hunting research progress. Only an exception is identification of orthologues of mammalian complement regulatory proteins and complement receptors. It is of particular interest that teleost complement components often exist as multiple isoforms with possible functional divergence. This review summarizes research progress of teleost complement system following the molecular identification and sequence analysis of the components. The findings of extensive expression analyses of the complement components with special emphasis of their prominent extrahepatic expression, acute-phase response to immunostimulation and various microbial infections, and ontogenic development including maternal transfer are discussed to infer teleost-specific functions of the complement system. Importance of the protein level characterization of the complement components is also emphasized, especially for understanding of the isotypic diversity of the components, a unique feature of teleost complement system.
Collapse
Affiliation(s)
- Miki Nakao
- Laboratory of Marine Biochemistry, Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 812-8581, Japan.
| | | | | | | | | |
Collapse
|
25
|
Neher MD, Weckbach S, Flierl MA, Huber-Lang MS, Stahel PF. Molecular mechanisms of inflammation and tissue injury after major trauma--is complement the "bad guy"? J Biomed Sci 2011; 18:90. [PMID: 22129197 PMCID: PMC3247859 DOI: 10.1186/1423-0127-18-90] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 11/30/2011] [Indexed: 02/07/2023] Open
Abstract
Trauma represents the leading cause of death among young people in industrialized countries. Recent clinical and experimental studies have brought increasing evidence for activation of the innate immune system in contributing to the pathogenesis of trauma-induced sequelae and adverse outcome. As the "first line of defense", the complement system represents a potent effector arm of innate immunity, and has been implicated in mediating the early posttraumatic inflammatory response. Despite its generic beneficial functions, including pathogen elimination and immediate response to danger signals, complement activation may exert detrimental effects after trauma, in terms of mounting an "innocent bystander" attack on host tissue. Posttraumatic ischemia/reperfusion injuries represent the classic entity of complement-mediated tissue damage, adding to the "antigenic load" by exacerbation of local and systemic inflammation and release of toxic mediators. These pathophysiological sequelae have been shown to sustain the systemic inflammatory response syndrome after major trauma, and can ultimately contribute to remote organ injury and death. Numerous experimental models have been designed in recent years with the aim of mimicking the inflammatory reaction after trauma and to allow the testing of new pharmacological approaches, including the emergent concept of site-targeted complement inhibition. The present review provides an overview on the current understanding of the cellular and molecular mechanisms of complement activation after major trauma, with an emphasis of emerging therapeutic concepts which may provide the rationale for a "bench-to-bedside" approach in the design of future pharmacological strategies.
Collapse
Affiliation(s)
- Miriam D Neher
- Department of Orthopaedic Surgery, University of Colorado Denver, School of Medicine, Denver Health Medical Center, 777 Bannock Street, Denver, CO 80204, USA
| | | | | | | | | |
Collapse
|
26
|
Human complement receptor type 2 (CR2/CD21) transgenic mice provide an in vivo model to study immunoregulatory effects of receptor antagonists. Mol Immunol 2011; 48:883-94. [DOI: 10.1016/j.molimm.2010.12.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 12/16/2010] [Accepted: 12/20/2010] [Indexed: 01/12/2023]
|
27
|
Marginal zone B cells are naturally reactive to collagen type II and are involved in the initiation of the immune response in collagen-induced arthritis. Cell Mol Immunol 2011; 8:296-304. [PMID: 21358667 DOI: 10.1038/cmi.2011.2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Antibodies against type II collagen (CII) are essential for development of collagen-induced arthritis (CIA), but how and where the B-cell response to CII is initiated is not fully known. We show here that naive DBA/1 mice display naturally reactive IgM and IgG anti-CII producing B cells prior to immunization. The CII-reactive B cells were observed in the spleen and recognized as marginal zone (MZ) B cells. After CII immunization, CII-specific B cells expanded rapidly in the spleen, in contrast to the lymph nodes, with the initial response derived from MZ B cells and later by follicular (FO) B cells. This was evident despite that the MZ B cells were subject to stringent tolerance mechanisms by having a greater Fc gamma receptor IIb expression than the FO B cells. Further, the MZ B cells migrated to the FO areas upon immunization, possibly providing antigen and activating FO T cells and subsequently FO B cells. Thus, around CIA onset increased numbers of IgG anti-CII producing FO B cells was seen in the spleen, which was dominated by IgG2a- and IgG2b-positive cells. These data demonstrate that CII-reactive MZ B cells are present before and expand after CII immunization, suggesting an initiating role of MZ B cells in the development of CIA.
Collapse
|
28
|
Woods KM, Pope MR, Hoffman SM, Fleming SD. CR2+ marginal zone B cell production of pathogenic natural antibodies is C3 independent. THE JOURNAL OF IMMUNOLOGY 2010; 186:1755-62. [PMID: 21187447 DOI: 10.4049/jimmunol.1002059] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intestinal ischemia-reperfusion (IR)-induced damage requires complement receptor 2 (CR2) for generation of the appropriate natural Ab repertoire. Pathogenic Abs recognize neoantigens on the ischemic tissue, activate complement, and induce intestinal damage. Because C3 cleavage products act as ligands for CR2, we hypothesized that CR2(hi) marginal zone B cells (MZBs) require C3 for generation of the pathogenic Abs. To explore the ability of splenic CR2(+) B cells to generate the damaging Ab repertoire, we adoptively transferred either MZBs or follicular B cells (FOBs) from C57BL/6 or Cr2(-/-) mice into Rag-1(-/-) mice. Adoptive transfer of wild type CR2(hi) MZBs but not CR2(lo) FOBs induced significant damage, C3 deposition, and inflammation in response to IR. In contrast, similarly treated Rag-1(-/-) mice reconstituted with either Cr2(-/-) MZB/B1 B cells (B1Bs) or FOBs lacked significant intestinal damage and displayed limited complement activation. To determine whether C3 cleavage products are critical in CR2-dependent Ab production, we evaluated the ability of the natural Ab repertoire of C3(-/-) mice to induce damage in response to IR. Infusion of C3(-/-) serum into Cr2(-/-) mice restored IR-induced tissue damage. Furthermore, Rag-1(-/-) mice sustained significant damage after infusion of Abs from C3(-/-) but not Cr2(-/-) mice. Finally, adoptive transfer of MZBs from C3(-/-) mice into Rag-1(-/-) mice resulted in significant tissue damage and inflammation. These data indicate that CR2 expression on MZBs is sufficient to induce the appropriate Abs required for IR-induced tissue damage and that C3 is not critical for generation of the pathogenic Abs.
Collapse
Affiliation(s)
- Keith M Woods
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | | | | | | |
Collapse
|
29
|
Fleming SD, Pope MR, Hoffman SM, Moses T, Bukovnik U, Tomich JM, Wagner LM, Woods KM. Domain V peptides inhibit beta2-glycoprotein I-mediated mesenteric ischemia/reperfusion-induced tissue damage and inflammation. THE JOURNAL OF IMMUNOLOGY 2010; 185:6168-78. [PMID: 20956350 DOI: 10.4049/jimmunol.1002520] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Reperfusion of ischemic tissue induces significant tissue damage in multiple conditions, including myocardial infarctions, stroke, and transplantation. Although not as common, the mortality rate of mesenteric ischemia/reperfusion (IR) remains >70%. Although complement and naturally occurring Abs are known to mediate significant damage during IR, the target Ags are intracellular molecules. We investigated the role of the serum protein, β2-glycoprotein I as an initiating Ag for Ab recognition and β2-glycoprotein I (β2-GPI) peptides as a therapeutic for mesenteric IR. The time course of β2-GPI binding to the tissue indicated binding and complement activation within 15 min postreperfusion. Treatment of wild-type mice with peptides corresponding to the lipid binding domain V of β2-GPI blocked intestinal injury and inflammation, including cellular influx and cytokine and eicosanoid production. The optimal therapeutic peptide (peptide 296) contained the lysine-rich region of domain V. In addition, damage and most inflammation were also blocked by peptide 305, which overlaps with peptide 296 but does not contain the lysine-rich, phospholipid-binding region. Importantly, peptide 296 retained efficacy after replacement of cysteine residues with serine. In addition, infusion of wild-type serum containing reduced levels of anti-β2-GPI Abs into Rag-1(-/-) mice prevented IR-induced intestinal damage and inflammation. Taken together, these data suggest that the serum protein β2-GPI initiates the IR-induced intestinal damage and inflammatory response and as such is a critical therapeutic target for IR-induced damage and inflammation.
Collapse
Affiliation(s)
- Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
The complement system consists of a tightly regulated network of proteins that play an important role in host defense and inflammation. Complement activation results in opsonization of pathogens and their removal by phagocytes, as well as cell lysis. Inappropriate complement activation and complement deficiencies are the underlying cause of the pathophysiology of many diseases such as systemic lupus erythematosus and asthma. This review represents an overview of the complement system in an effort to understand the beneficial as well as harmful roles it plays during inflammatory responses.
Collapse
Affiliation(s)
- J Vidya Sarma
- Department of Pathology, The University of Michigan Medical School, 1301 Catherine Rd., Box 5602, Ann Arbor, MI 48109-5602, USA
| | | |
Collapse
|
31
|
Dunkelberger JR, Song WC. Role and mechanism of action of complement in regulating T cell immunity. Mol Immunol 2010; 47:2176-86. [PMID: 20603023 DOI: 10.1016/j.molimm.2010.05.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Complement is a part of the innate immune system that contributes to first-line host defense. It is also implicated in a number of human inflammatory conditions and has attracted interest as a potential therapeutic target. Understanding the basic biology of complement and its mechanism(s) of action is imperative for developing complement-based treatments for infectious and autoimmune diseases. One of the exciting new developments in this regard is the revelation that complement plays an important role in T cell immunity. In this review, we highlight recent published studies implicating complement in models of CD4+ and CD8+ T cell immune responses, and discuss its potential mechanism(s) action in these processes. We also comment on issues that may impact data interpretation and draw attention to their consideration in future studies.
Collapse
Affiliation(s)
- Jason R Dunkelberger
- Institute for Translational Medicine and Therapeutics and Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
32
|
Bergström FC, Reynolds S, Johnstone M, Pike RN, Buckle AM, Kemp DJ, Fischer K, Blom AM. Scabies mite inactivated serine protease paralogs inhibit the human complement system. THE JOURNAL OF IMMUNOLOGY 2009; 182:7809-17. [PMID: 19494305 DOI: 10.4049/jimmunol.0804205] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Infestation of skin by the parasitic itch mite Sarcoptes scabiei afflicts 300 million people worldwide and there is a need for novel and efficient therapies. We have previously identified a multigene family of serine proteases comprising multiple catalytically inactive members (scabies mite-inactivated protease paralogs (SMIPPs)), which are secreted into the gut of S. scabiei. SMIPPs are located in the mite gut and in feces excreted into the upper epidermis. Scabies mites feed on epidermal protein, including host plasma; consequently, they are exposed to host defense mechanisms both internally and externally. We found that two recombinantly expressed SMIPPs inhibited all three pathways of the human complement system. Both SMIPPs exerted their inhibitory action due to binding of three molecules involved in the three different mechanisms which initiate complement: C1q, mannose-binding lectin, and properdin. Both SMIPPs bound to the stalk domains of C1q, possibly displacing or inhibiting C1r/C1s, which are associated with the same domain. Furthermore, we found that binding of both SMIPPs to properdin resulted in prevention of assembly of the alternative pathway convertases. However, the SMIPPs were not able to dissociate already formed convertases. Immunohistochemical staining demonstrated the presence of C1q in the gut of scabies mites in skin burrows. We propose that SMIPPs minimize complement-mediated gut damage and thus create a favorable environment for the scabies mites.
Collapse
Affiliation(s)
- Frida C Bergström
- Department of Laboratory Medicine, Wallenberg Laboratory, University Hospital Malmö, Lund University, Malmö, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Seregin SS, Aldhamen YA, Appledorn DM, Schuldt NJ, McBride AJ, Bujold M, Godbehere SS, Amalfitano A. CR1/2 is an important suppressor of Adenovirus-induced innate immune responses and is required for induction of neutralizing antibodies. Gene Ther 2009; 16:1245-59. [PMID: 19554032 PMCID: PMC4039027 DOI: 10.1038/gt.2009.77] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Human complement receptors 1 and 2 are well described as important regulators of innate and adaptive immune responses, having pivotal roles in regulating complement activation (CR1) and B cell maturation/survival. In contrast, the role of the murine homologues of CR1 and CR2 (mCR1/2) have been primarily defined as modulating activation of the adaptive immune system, with very little evidence available about the role of mCR1/2 in regulating the innate immune responses to pathogens. In this manuscript, we confirm that mCR1/2 plays an important role in regulating both the innate and adaptive immune responses noted after Adenovirus (Ad) mediated gene transfer. Our results uncovered a novel role of mCR1/2 in down-regulating several, complement dependent innate immune responses. We also unveiled the mechanism underlying the complement dependent induction of neutralizing antibodies to Ad capsids as a CR1/2 dependent phenomenon that correlates with B-cell activation. These results confirm that Ad interactions with the complement system are pivotal in understanding how to maximize the safety or potency of Ad mediated gene transfer for both gene therapy and vaccine applications.
Collapse
Affiliation(s)
- S S Seregin
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Kulik L, Fleming SD, Moratz C, Reuter JW, Novikov A, Chen K, Andrews KA, Markaryan A, Quigg RJ, Silverman GJ, Tsokos GC, Holers VM. Pathogenic natural antibodies recognizing annexin IV are required to develop intestinal ischemia-reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2009; 182:5363-73. [PMID: 19380783 DOI: 10.4049/jimmunol.0803980] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Intestinal ischemia-reperfusion (IR) injury is initiated when natural IgM Abs recognize neo-epitopes that are revealed on ischemic cells. The target molecules and mechanisms whereby these neo-epitopes become accessible to recognition are not well understood. Proposing that isolated intestinal epithelial cells (IEC) may carry IR-related neo-epitopes, we used in vitro IEC binding assays to screen hybridomas created from B cells of unmanipulated wild-type C57BL/6 mice. We identified a novel IgM mAb (mAb B4) that reacted with the surface of IEC by flow cytometric analysis and was alone capable of causing complement activation, neutrophil recruitment and intestinal injury in otherwise IR-resistant Rag1(-/-) mice. mAb B4 was found to specifically recognize mouse annexin IV. Preinjection of recombinant annexin IV blocked IR injury in wild-type C57BL/6 mice, demonstrating the requirement for recognition of this protein to develop IR injury in the context of a complex natural Ab repertoire. Humans were also found to exhibit IgM natural Abs that recognize annexin IV. These data in toto identify annexin IV as a key ischemia-related target Ag that is recognized by natural Abs in a pathologic process required in vivo to develop intestinal IR injury.
Collapse
Affiliation(s)
- Liudmila Kulik
- Department of Medicine, University of Colorado Denver School of Medicine, Denver, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Molecular structure and expression of anthropic, ovine, and murine forms of complement receptor type 2. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 15:901-10. [PMID: 18400970 DOI: 10.1128/cvi.00465-07] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
36
|
Atkinson C, Qiao F, Song H, Gilkeson GS, Tomlinson S. Low-dose targeted complement inhibition protects against renal disease and other manifestations of autoimmune disease in MRL/lpr mice. THE JOURNAL OF IMMUNOLOGY 2008; 180:1231-8. [PMID: 18178863 DOI: 10.4049/jimmunol.180.2.1231] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Complement appears to play a dual role in the progression of systemic lupus erythematosus, serving a beneficial role in enhancing immune complex clearance, while serving a pathogenic role in inducing local inflammation. To investigate these different roles of complement in a therapeutic setting, MRL/lpr mice were treated with the targeted murine C3 complement inhibitor, CR2-Crry, from 16 to 24 wk of age (after the development of proteinuria). The targeting moiety, CR2, binds to C3 breakdown products deposited at sites of complement activation and has the potential to provide complement inhibition locally without causing systemic inhibition. Administration of CR2-Crry i.v., at a dose of 0.25 mg once a week, was associated with a significant survival benefit, improved kidney function, and a significant reduction in glomerulonephritis and renal vasculitis. The presence of skin lesions and lung bronchiolar and vascular inflammation was also dramatically reduced by CR2-Crry treatment. CR2-Crry treatment also resulted in a significant reduction in autoantibody production, as measured by anti-dsDNA Ab levels, and did not cause an increase in circulating immune complex levels. These effects on autoimmunity and circulating immune complexes represent significant potential advantages over the use of Crry-Ig in MRL/lpr mice, a systemic counterpart of CR2-Crry. CR2-Crry localized preferentially to the kidneys in 16-wk MRL/lpr mice with a kidney-localized half-life of approximately 24 h. Thus, targeted complement inhibition at the C3 level is an effective treatment in murine lupus, even beginning after onset of disease.
Collapse
Affiliation(s)
- Carl Atkinson
- Department of Microbiology and Immunology and Children's Research Institute, Medical University ofSouth Carolina, Charleston 29425, USA
| | | | | | | | | |
Collapse
|
37
|
Transcriptional control of complement receptor gene expression. Immunol Res 2008; 39:146-59. [PMID: 17917062 DOI: 10.1007/s12026-007-0078-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 02/01/2023]
Abstract
Immune complement is a critical system in the immune response and protection of host cells from damage by complement is critical during inflammation. The expression of the receptors for the inflammatory anaphylatoxin molecules is also key in immunity. In order to fully appreciate the biology of complement, a basic understanding of the molecular regulation of complement receptor gene expression is critical, yet these kinds of studies are lacking for many genes. Importantly, recent genetic studies have demonstrated that promoter-enhancer polymorphisms can contribute to pathology in diseases such as atypical hemolytic uremic syndrome. This review will focus on what is currently known about the genetic regulation of key protective complement receptors genes including CR1 (CD35), CR2 (CD21), Crry, MCP (CD46), DAF (CD55), and CD59. In addition, the regulation of the anaphylatoxin receptors genes, C3aR and C5aR (CD88) will also be discussed. Since new research continuously uncovers novel functions for these proteins, a greater appreciation of the mechanisms involved in gene regulation will be critical for understanding the biology of these molecules.
Collapse
|
38
|
Abstract
The complement system is an important component of the innate immune system and a modulator of adaptive immunity. The entire complement system is focused on C3 and C5. Thus, there are proteins that activate C3 and C5, those that regulate this activation, and those that transduce the effects of C3 and C5 activation products; each can affect the kidney in renal injury. The normal kidney has the inherent capacity to protect itself from complement activation through cellular expression of decay-accelerating factor, membrane cofactor protein (in human beings), and Crry (in rodents). In addition, plasma factor H protects vascular spaces in the kidney. Although the main function of these proteins is to limit complement activation, there is now considerable evidence that they can transduce signals on engagement in immune cells. The G-protein-coupled 7-span transmembrane receptors for C3a and C5a, and the integral membrane complement receptors (CR) for C3b, iC3b, and C3dg, are expressed outside the kidney, particularly in cells of hematopoietic and immune lineage. These are important in renal injury through their infiltration of the kidney and/or by affecting kidney-directed immune responses. There is mounting evidence that intrinsic glomerular and tubular cell C3aR and C5aR expression and activation also can affect renal injury. CR1 on podocytes and the beta2 integrins CR3 and CR4 in kidney dendritic cells have functions that remain poorly defined. Cells of the kidney also have the capacity to produce and activate their own complement proteins. Thus, intrinsic renal cells express decay-accelerating factor, membrane cofactor protein, Crry, C3aR, C5aR, CR1, CR3, and CR4. These can be engaged by C3 and C5 activation products derived from systemic and local pools in renal injury. Given their capacity to provide signals that influence kidney cellular behavior, their activation can have substantial effects in renal injury. Defining these in a cell- and disease-specific fashion is an exciting challenge for future research.
Collapse
Affiliation(s)
- Tipu S Puri
- Section of Nephrology, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
39
|
|