1
|
Holers VM. Complement therapeutics are coming of age in rheumatology. Nat Rev Rheumatol 2023; 19:470-485. [PMID: 37337038 DOI: 10.1038/s41584-023-00981-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 06/21/2023]
Abstract
The complement system was described over 100 years ago, and it is well established that activation of this pathway accompanies the great majority of autoimmune and inflammatory diseases. In addition, over three decades of work in murine models of human disease have nearly universally demonstrated that complement activation is upstream of tissue injury and the engagement of pro-inflammatory mechanisms such as the elaboration of cytokines and chemokines, as well as myeloid cell recruitment and activation. With that background, and taking advantage of advances in the development of biologic and small-molecule therapeutics, the creation and clinical evaluation of complement therapeutics is now rapidly expanding. This article reviews the current state of the complement therapeutics field, with a focus on their use in diseases cared for or consulted upon by rheumatologists. Included is an overview of the activation mechanisms and components of the system, in addition to the mechanisms by which the complement system interacts with other immune system constituents. The various therapeutic approaches to modulating the system in rheumatic and autoimmune diseases are reviewed. To understand how best to clinically assess the complement system, methods of its evaluation are described. Finally, next-generation therapeutic and diagnostic advances that can be envisioned for the future are discussed.
Collapse
Affiliation(s)
- V Michael Holers
- Medicine/Rheumatology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
2
|
Sun XY, Yu XL, Zhu J, Li LJ, Zhang L, Huang YR, Liu DQ, Ji M, Sun X, Zhang LX, Zhou WW, Zhang D, Jiao J, Liu RT. Fc effector of anti-Aβ antibody induces synapse loss and cognitive deficits in Alzheimer's disease-like mouse model. Signal Transduct Target Ther 2023; 8:30. [PMID: 36693826 PMCID: PMC9873795 DOI: 10.1038/s41392-022-01273-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 01/26/2023] Open
Abstract
Passive immunotherapy is one of the most promising interventions for Alzheimer's disease (AD). However, almost all immune-modulating strategies fail in clinical trials with unclear causes although they attenuate neuropathology and cognitive deficits in AD animal models. Here, we showed that Aβ-targeting antibodies including their lgG1 and lgG4 subtypes induced microglial engulfment of neuronal synapses by activating CR3 or FcγRIIb via the complex of Aβ, antibody, and complement. Notably, anti-Aβ antibodies without Fc fragment, or with blockage of CR3 or FcγRIIb, did not exert these adverse effects. Consistently, Aβ-targeting antibodies, but not their Fab fragments, significantly induced acute microglial synapse removal and rapidly exacerbated cognitive deficits and neuroinflammation in APP/PS1 mice post-treatment, whereas the memory impairments in mice were gradually rescued thereafter. Since the recovery rate of synapses in humans is much lower than that in mice, our findings may clarify the variances in the preclinical and clinical studies assessing AD immunotherapies. Therefore, Aβ-targeting antibodies lack of Fc fragment, or with reduced Fc effector function, may not induce microglial synaptic pruning, providing a safer and more efficient therapeutic alternative for passive immunotherapy for AD.
Collapse
Affiliation(s)
- Xiao-ying Sun
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China ,grid.410726.60000 0004 1797 8419School of Chemistry and Chemical Engineering, University of Chinese Academy of Science, Beijing, 100049 China
| | - Xiao-lin Yu
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China ,grid.9227.e0000000119573309Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190 China
| | - Jie Zhu
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China ,grid.410726.60000 0004 1797 8419School of Chemistry and Chemical Engineering, University of Chinese Academy of Science, Beijing, 100049 China
| | - Ling-jie Li
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China ,grid.410726.60000 0004 1797 8419School of Chemistry and Chemical Engineering, University of Chinese Academy of Science, Beijing, 100049 China
| | - Lun Zhang
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China ,grid.9227.e0000000119573309Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190 China
| | - Ya-ru Huang
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China ,grid.410726.60000 0004 1797 8419School of Chemistry and Chemical Engineering, University of Chinese Academy of Science, Beijing, 100049 China
| | - Dong-qun Liu
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China
| | - Mei Ji
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China
| | - Xun Sun
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China
| | - Ling-xiao Zhang
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China
| | - Wei-wei Zhou
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China ,grid.9227.e0000000119573309Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190 China
| | - Dongming Zhang
- grid.9227.e0000000119573309State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Jianwei Jiao
- grid.9227.e0000000119573309State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China
| | - Rui-tian Liu
- grid.9227.e0000000119573309State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China ,grid.9227.e0000000119573309Innovation Academy for Green Manufacture Institute, Chinese Academy of Sciences, Beijing, 100190 China
| |
Collapse
|
3
|
Holers VM. Contributions of animal models to mechanistic understandings of antibody-dependent disease and roles of the amplification loop. Immunol Rev 2023; 313:181-193. [PMID: 36111456 DOI: 10.1111/imr.13136] [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] [Indexed: 01/20/2023]
Abstract
The complement system plays an important pathophysiologic role in human diseases associated with immune or ischemic insults. In addition to understanding the effector mechanisms that are important for the biological effects of the system, substantial efforts have gone into understanding which specific complement activation pathways generate these potent effects. These approaches include the use of gene-targeted mice and specific pathway inhibitors, as well as the integration of human disease genetic and biomarker studies. In some disease states, it is quite clear that the alternate pathway plays a unique role in the initiation of the complement system. However, although initially a widely unexpected finding, it has now been shown in many tissue-based disease models and in initial human studies that engagement of the amplification loop is also essential for tissue injury when the classical and/or lectin pathways initiate pathway activation through pathogenic autoantibodies. This review provides evidence for such a conclusion through animal models, focusing on pathogenic antibody passive transfer models but also other relevant experimental systems. These data, along with initial biomarkers and clinical trial outcomes in human diseases that are associated with pathogenic autoantibodies, suggest that targeting the alternative pathway amplification loop may have near-universal therapeutic utility for tissue-based diseases.
Collapse
Affiliation(s)
- V Michael Holers
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
4
|
Chen K, Deng Y, Shang S, Tang L, Li Q, Bai X, Chen X. Complement factor B inhibitor LNP023 improves lupus nephritis in MRL/lpr mice. Biomed Pharmacother 2022; 153:113433. [DOI: 10.1016/j.biopha.2022.113433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
|
5
|
Pryzdial ELG, Leatherdale A, Conway EM. Coagulation and complement: Key innate defense participants in a seamless web. Front Immunol 2022; 13:918775. [PMID: 36016942 PMCID: PMC9398469 DOI: 10.3389/fimmu.2022.918775] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/06/2022] [Indexed: 12/30/2022] Open
Abstract
In 1969, Dr. Oscar Ratnoff, a pioneer in delineating the mechanisms by which coagulation is activated and complement is regulated, wrote, “In the study of biological processes, the accumulation of information is often accelerated by a narrow point of view. The fastest way to investigate the body’s defenses against injury is to look individually at such isolated questions as how the blood clots or how complement works. We must constantly remind ourselves that such distinctions are man-made. In life, as in the legal cliché, the devices through which the body protects itself form a seamless web, unwrinkled by our artificialities.” Our aim in this review, is to highlight the critical molecular and cellular interactions between coagulation and complement, and how these two major component proteolytic pathways contribute to the seamless web of innate mechanisms that the body uses to protect itself from injury, invading pathogens and foreign surfaces.
Collapse
Affiliation(s)
- Edward L. G. Pryzdial
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Canadian Blood Services, Medical Affairs and Innovation, Vancouver, BC, Canada
- *Correspondence: Edward L. G. Pryzdial, ; Edward M. Conway,
| | - Alexander Leatherdale
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Edward M. Conway
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Canadian Blood Services, Medical Affairs and Innovation, Vancouver, BC, Canada
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Edward L. G. Pryzdial, ; Edward M. Conway,
| |
Collapse
|
6
|
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: 1.0] [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
|
7
|
Affiliation(s)
- Hayeong Rho
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Richard A Wells
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Canada
- Division of Medical Oncology and Hematology, Odette Cancer Centre, Sunnybrook Health Science Centre, Toronto, Canada
| |
Collapse
|
8
|
Natali EN, Principato S, Ferlicca F, Bianchi F, Fontana LE, Faleri A, Pansegrau W, Surdo PL, Bartolini E, Santini L, Brunelli B, Giusti F, Veggi D, Ferlenghi I, Norais N, Scarselli M. Synergic complement-mediated bactericidal activity of monoclonal antibodies with distinct specificity. FASEB J 2020; 34:10329-10341. [PMID: 32725956 DOI: 10.1096/fj.201902795r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 04/30/2020] [Accepted: 05/20/2020] [Indexed: 01/24/2023]
Abstract
The classical complement pathway is triggered when antigen-bound immunoglobulins bind to C1q through their Fc region. While C1q binds to a single Fc with low affinity, a higher avidity stable binding of two or more of C1q globular heads initiates the downstream reactions of the complement cascade ultimately resulting in bacteriolysis. Synergistic bactericidal activity has been demonstrated when monoclonal antibodies recognize nonoverlapping epitopes of the same antigen. The aim of the present work was to investigate the synergistic effect between antibodies directed toward different antigens. To this purpose, we investigated the bactericidal activity induced by combinations of monoclonal antibodies (mAbs) raised against factor H-binding protein (fHbp) and Neisserial Heparin-Binding Antigen (NHBA), two major antigens included in Bexsero, the vaccine against Meningococcus B, for prevention from this devastating disease in infants and adolescents. Collectively, our results show that mAbs recognizing different antigens can synergistically activate complement even when each single Mab is not bactericidal, reinforcing the evidence that cooperative immunity induced by antigen combinations can represent a remarkable added value of multicomponent vaccines. Our study also shows that the synergistic effect of antibodies is modulated by the nature of the respective epitopes, as well as by the antigen density on the bacterial cell surface.
Collapse
Affiliation(s)
- Eriberto Noel Natali
- GSK, Siena, Italy.,CERM, Department of Chemistry, University of Florence, Florence, Italy
| | - Silvia Principato
- GSK, Siena, Italy.,Department of Biological Sciences, University of Siena, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Mainolfi N, Ehara T, Karki RG, Anderson K, Mac Sweeney A, Liao SM, Argikar UA, Jendza K, Zhang C, Powers J, Klosowski DW, Crowley M, Kawanami T, Ding J, April M, Forster C, Serrano-Wu M, Capparelli M, Ramqaj R, Solovay C, Cumin F, Smith TM, Ferrara L, Lee W, Long D, Prentiss M, De Erkenez A, Yang L, Liu F, Sellner H, Sirockin F, Valeur E, Erbel P, Ostermeier D, Ramage P, Gerhartz B, Schubart A, Flohr S, Gradoux N, Feifel R, Vogg B, Wiesmann C, Maibaum J, Eder J, Sedrani R, Harrison RA, Mogi M, Jaffee BD, Adams CM. Discovery of 4-((2 S,4 S)-4-Ethoxy-1-((5-methoxy-7-methyl-1 H-indol-4-yl)methyl)piperidin-2-yl)benzoic Acid (LNP023), a Factor B Inhibitor Specifically Designed To Be Applicable to Treating a Diverse Array of Complement Mediated Diseases. J Med Chem 2020; 63:5697-5722. [PMID: 32073845 DOI: 10.1021/acs.jmedchem.9b01870] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The alternative pathway (AP) of the complement system is a key contributor to the pathogenesis of several human diseases including age-related macular degeneration, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and various glomerular diseases. The serine protease factor B (FB) is a key node in the AP and is integral to the formation of C3 and C5 convertase. Despite the prominent role of FB in the AP, selective orally bioavailable inhibitors, beyond our own efforts, have not been reported previously. Herein we describe in more detail our efforts to identify FB inhibitors by high-throughput screening (HTS) and leveraging insights from several X-ray cocrystal structures during optimization efforts. This work culminated in the discovery of LNP023 (41), which is currently being evaluated clinically in several diverse AP mediated indications.
Collapse
Affiliation(s)
- Nello Mainolfi
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Takeru Ehara
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Rajeshri G Karki
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Karen Anderson
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Aengus Mac Sweeney
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Sha-Mei Liao
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Upendra A Argikar
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Keith Jendza
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Chun Zhang
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - James Powers
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Daniel W Klosowski
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Maura Crowley
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Toshio Kawanami
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Jian Ding
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Myriam April
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Cornelia Forster
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Michael Serrano-Wu
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Michael Capparelli
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Rrezarta Ramqaj
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Catherine Solovay
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Frederic Cumin
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Thomas M Smith
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Luciana Ferrara
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Wendy Lee
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Debby Long
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Melissa Prentiss
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Andrea De Erkenez
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Louis Yang
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Fang Liu
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Holger Sellner
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Finton Sirockin
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Eric Valeur
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Paulus Erbel
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Daniela Ostermeier
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Paul Ramage
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Bernd Gerhartz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Anna Schubart
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Stefanie Flohr
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Nathalie Gradoux
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Roland Feifel
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Barbara Vogg
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Christian Wiesmann
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Jürgen Maibaum
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Jörg Eder
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Richard Sedrani
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Richard A Harrison
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Muneto Mogi
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Bruce D Jaffee
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| | - Christopher M Adams
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
10
|
Ramakrishnan P, Nagarajan D. Neuromyelitis optica spectrum disorder: an overview. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
11
|
Kim H, Conway EM. Platelets and Complement Cross-Talk in Early Atherogenesis. Front Cardiovasc Med 2019; 6:131. [PMID: 31555668 PMCID: PMC6742699 DOI: 10.3389/fcvm.2019.00131] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 08/22/2019] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis remains a ubiquitous and serious threat to human health. The initial formation of the atherosclerotic lesion (atheroma) is driven by pro-inflammatory signaling involving monocytes and vascular endothelial cells; later stages of the disease involve rupture of well-established atherosclerotic plaques, thrombosis, and blood vessel occlusion. While the central role of platelets in thrombosis is undisputed, platelets exhibit pro-inflammatory activities, and contribute to early-stage atheroma formation. Platelets also engage components of the complement system, an essential element of innate immunity that contributes to vascular inflammation. Here we provide an overview of the complex interplay between platelets and the complement system, with a focus on how the crosstalk between them may impact on the initiation of atheroma formation.
Collapse
Affiliation(s)
- Hugh Kim
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.,Faculty of Dentistry, University of British Columbia, Vancouver, BC, Canada
| | - Edward M Conway
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
12
|
Savelli SL, Roubey RAS, Kitzmiller KJ, Zhou D, Nagaraja HN, Mulvihill E, Barbar-Smiley F, Ardoin SP, Wu YL, Yu CY. Opposite Profiles of Complement in Antiphospholipid Syndrome (APS) and Systemic Lupus Erythematosus (SLE) Among Patients With Antiphospholipid Antibodies (aPL). Front Immunol 2019; 10:885. [PMID: 31134052 PMCID: PMC6514053 DOI: 10.3389/fimmu.2019.00885] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/05/2019] [Indexed: 12/18/2022] Open
Abstract
APS is the association of antiphospholipid antibodies (aPL) with thromboses and/or recurrent pregnancy loss (RPL). Among patients with SLE, one-third have aPL and 10–15% have a manifestation of secondary APS. Animal studies suggested that complement activation plays an important role in the pathogenesis of thrombosis and pregnancy loss in APS. We performed a cross-sectional study on complement proteins and genes in 525 patients with aPL. Among them, 237 experienced thromboses and 293 had SLE; 111 had both SLE and thromboses, and 106 had neither SLE nor thrombosis. Complement protein levels were determined by radial immunodiffusion for C4, C3 and factor H; and by functional ELISA for mannan binding lectin (MBL). Total C4, C4A and C4B gene copy numbers (GCN) were measured by TaqMan-based realtime PCR. Two to six copies of C4 genes are frequently present in a diploid genome, and each copy may code for an acidic C4A or a basic C4B protein. We observed significantly (a) higher protein levels of total C4, C4A, C4B, C3, and anticardiolipin (ACLA) IgG, (b) increased frequencies of lupus anticoagulant and males, and (c) decreased levels of complement factor H, MBL and ACLA-IgM among patients with thrombosis than those without thrombosis (N = 288). We also observed significantly lower GCNs of total C4 and C4A among aPL-positive patients with both SLE and thrombosis than others. By contrast, aPL-positive subjects with SLE had significantly reduced protein levels of C3, total C4, C4A, C4B and ACLA-IgG, and higher frequency of females than those without SLE. Patients with thrombosis but without SLE (N = 126), and patients with SLE but without thrombosis (N = 182) had the greatest differences in mean protein levels of C3 (p = 2.6 × 10−6), C4 (p = 2.2 × 10−9) and ACLA-IgG (p = 1.2 × 10−5). RPL occurred in 23.7% of female patients and thrombotic SLE patients had the highest frequency of RPL (41.0%; p = 3.8 × 10−10). Compared with non-RPL females, RPL had significantly higher frequency of thrombosis and elevated C4 protein levels. Female patients with homozygous C4A deficiency all experienced RPL (p = 0.0001) but the opposite was true for patients with homozygous C4B deficiency (p = 0.017). These results provide new insights and biomarkers for diagnosis and management of APS and SLE.
Collapse
Affiliation(s)
- Stephanie L Savelli
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Division of Hematology/Oncology, Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Robert A S Roubey
- Division of Rheumatology, Allergy and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kathryn J Kitzmiller
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Danlei Zhou
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States.,Division of Rheumatology, Nationwide Children's Hospital, Columbus, OH, United States
| | - Haikady N Nagaraja
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH, United States
| | - Evan Mulvihill
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States.,Division of Rheumatology, Nationwide Children's Hospital, Columbus, OH, United States
| | - Fatima Barbar-Smiley
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States.,Division of Rheumatology, Nationwide Children's Hospital, Columbus, OH, United States
| | - Stacy P Ardoin
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States.,Division of Rheumatology, Nationwide Children's Hospital, Columbus, OH, United States
| | - Yee Ling Wu
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States.,Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, United States
| | - Chack-Yung Yu
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States.,Division of Rheumatology, Nationwide Children's Hospital, Columbus, OH, United States
| |
Collapse
|
13
|
Soltys J, Liu Y, Ritchie A, Wemlinger S, Schaller K, Schumann H, Owens GP, Bennett JL. Membrane assembly of aquaporin-4 autoantibodies regulates classical complement activation in neuromyelitis optica. J Clin Invest 2019; 129:2000-2013. [PMID: 30958797 DOI: 10.1172/jci122942] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 02/26/2019] [Indexed: 01/29/2023] Open
Abstract
Neuromyelitis optica (NMO) is an autoimmune CNS disorder mediated by pathogenic aquaporin-4 (AQP4) water channel autoantibodies (AQP4-IgG). Although AQP4-IgG-driven complement-dependent cytotoxicity (CDC) is critical for the formation of NMO lesions, the molecular mechanisms governing optimal classical pathway activation are unknown. We investigated the molecular determinants driving CDC in NMO using recombinant AQP4-specific autoantibodies (AQP4 rAbs) derived from affected patients. We identified a group of AQP4 rAbs targeting a distinct extracellular loop C epitope that demonstrated enhanced CDC on target cells. Targeted mutations of AQP4 rAb Fc domains that enhance or diminish C1q binding or antibody Fc-Fc interactions showed that optimal CDC was driven by the assembly of multimeric rAb platforms that increase multivalent C1q binding and facilitate C1q activation. A peptide that blocks antibody Fc-Fc interaction inhibited CDC induced by AQP4 rAbs and polyclonal NMO patient sera. Super-resolution microscopy revealed that AQP4 rAbs with enhanced CDC preferentially formed organized clusters on supramolecular AQP4 orthogonal arrays, linking epitope-dependent multimeric assembly with enhanced C1q binding and activation. The resulting model of AQP4-IgG CDC provides a framework for understanding classical complement activation in human autoantibody-mediated disorders and identifies a potential new therapeutic avenue for treating NMO.
Collapse
Affiliation(s)
- John Soltys
- Neuroscience and Medical Scientist Training Programs
| | | | | | | | | | | | | | - Jeffrey L Bennett
- Neuroscience and Medical Scientist Training Programs.,Department of Neurology, and.,Department of Ophthalmology, University of Colorado at Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
14
|
Conway EM. Polyphosphates and Complement Activation. Front Med (Lausanne) 2019; 6:67. [PMID: 31019911 PMCID: PMC6458250 DOI: 10.3389/fmed.2019.00067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/18/2019] [Indexed: 01/11/2023] Open
Abstract
To sustain life in environments that are fraught with risks of life-threatening injury, organisms have developed innate protective strategies such that the response to wounds is rapid and localized, with the simultaneous recruitment of molecular, biochemical, and cellular pathways that limit bleeding and eliminate pathogens and damaged host cells, while promoting effective healing. These pathways are both coordinated and tightly regulated, as their over- or under-activation may lead to inadequate healing, disease, and/or demise of the host. Recent advances in our understanding of coagulation and complement, a key component of innate immunity, have revealed an intriguing linkage of the two systems. Cell-secreted polyphosphate promotes coagulation, while dampening complement activation, discoveries that are providing insights into disease mechanisms and suggesting novel therapeutic strategies.
Collapse
Affiliation(s)
- Edward M Conway
- Division of Hematology, Department of Medicine, Faculty of Medicine, Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Abstract
Complement is a key component of the innate immune system; however, its dysregulation due to genetic mutations or the presence of autoantibodies can cause a number of diseases including age-related macular degeneration, paroxysmal nocturnal hemoglobinuria, and C3 glomerulopathy. The alternative pathway acts as an amplification loop of the complement system. It is triggered by the activation of factor B, the proteolytically active component of the C3 and C5 convertases. We report the discovery of a small-molecule inhibitor of factor B and demonstrate that it can efficiently block the alternative pathway in vivo in animals and ex vivo in patient samples. The compound is highly selective and potent and is currently in clinical development for a number of complement-mediated diseases. Dysregulation of the alternative complement pathway (AP) predisposes individuals to a number of diseases including paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, and C3 glomerulopathy. Moreover, glomerular Ig deposits can lead to complement-driven nephropathies. Here we describe the discovery of a highly potent, reversible, and selective small-molecule inhibitor of factor B, a serine protease that drives the central amplification loop of the AP. Oral administration of the inhibitor prevents KRN-induced arthritis in mice and is effective upon prophylactic and therapeutic dosing in an experimental model of membranous nephropathy in rats. In addition, inhibition of factor B prevents complement activation in sera from C3 glomerulopathy patients and the hemolysis of human PNH erythrocytes. These data demonstrate the potential therapeutic value of using a factor B inhibitor for systemic treatment of complement-mediated diseases and provide a basis for its clinical development.
Collapse
|
16
|
Abstract
: Complement and coagulation are evolutionarily related proteolytic cascades in the blood that are critical for effecting an appropriate innate response to injury that limits bleeding and infection, while promoting healing. Although often viewed as distinct, it has long been recognized that cross-talk likely exists between these pathways. Only recently have molecular links been established. These are providing insights that are revealing opportunities for the development of novel therapeutic strategies to better treat a wide range of thrombotic, inflammatory, immune, infectious, and malignant diseases. In this brief review, the complex relationship between complement and coagulation is highlighted, underlining some of the newly uncovered interactions, in the hopes of stimulating innovative research that will yield improvements in patient outcomes.
Collapse
|
17
|
The effect on the immunology laboratory of the expansion in complement therapeutics. J Immunol Methods 2018; 461:30-36. [PMID: 30092178 DOI: 10.1016/j.jim.2018.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 12/11/2022]
Abstract
The approval in the US and Europe of Eculizumab in 2007 marked a change in complement therapeutics, and with it the landscape for complement testing in the clinical immunology laboratory changed. The change had begun even before that when C1-Inhibitor preparations were approved in the 1980s in Europe. There are now two classes of approved drugs that may impact the immunology laboratory, with two dozen more with novel modalities and potential indications that are in various stages of development. Every pathway and about every component of complement has been targeted by these drug development programs, and the modalities of the drugs in development are diverse. These developments will likely result in more laboratories offering more complement testing, so this review looks forward to some of those possible changes in testing.
Collapse
|
18
|
Sartain SE, Turner NA, Moake JL. Brain microvascular endothelial cells exhibit lower activation of the alternative complement pathway than glomerular microvascular endothelial cells. J Biol Chem 2018; 293:7195-7208. [PMID: 29555686 DOI: 10.1074/jbc.ra118.002639] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Indexed: 12/20/2022] Open
Abstract
Atypical hemolytic uremic syndrome (aHUS) and bone marrow transplantation-associated thrombotic microangiopathy (TA-TMA) are associated with excessive activation of the alternative complement pathway (AP) and with severe renal, but rarely cerebral, microvascular damage. Here, we compared AP activation and regulation in human glomerular and brain microvascular endothelial cells (GMVECs and BMVECs, respectively) unstimulated or stimulated by the proinflammatory cytokine, tumor necrosis factor (TNF). Compared with GMVECs and under both experimental conditions, BMVECs had increased gene expression of the AP-related genes C3, CFB, and C5 and decreased expression of CFD This was associated with increased expression in BMVECs (relative to GMVECs) of the genes for surface and soluble regulatory molecules (CD46, THBD, CD55, CFI, and CFH) suppressing formation of the AP C3 and C5 convertases. Of note, unlike GMVECs, BMVECs generated extremely low levels of C3a and C5a and displayed decreased activation of the AP (as measured by a lower percentage of Ba generation than GMVECs). Moreover, BMVECs exhibited increased function of CD141, mediating activation of the natural anticoagulant protein C, compared with GMVECs. We also found that the C3a receptor (C3aR) is present on both cell types and that TNF greatly increases C3AR1 expression in GMVECs, but only slightly in BMVECs. Higher AP activation and C3a generation in GMVECs than in BMVECs, coupled with an increase in C3aR production in TNF-stimulated GMVECs, provides a possible explanation for the predominance of renal damage, and the absence of cerebral injury, in individuals with episodes of aHUS and TA-TMA.
Collapse
Affiliation(s)
- Sarah E Sartain
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas 77030.
| | - Nancy A Turner
- Department of Bioengineering, Rice University, Houston, Texas 77005
| | - Joel L Moake
- Department of Bioengineering, Rice University, Houston, Texas 77005
| |
Collapse
|
19
|
Affiliation(s)
- Lucio Luzzatto
- From the Department of Hematology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania (L.L.); and the Department of Oncology, Biochemistry Unit, University of Turin, Turin, Italy (P.A.)
| | - Paolo Arese
- From the Department of Hematology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania (L.L.); and the Department of Oncology, Biochemistry Unit, University of Turin, Turin, Italy (P.A.)
| |
Collapse
|
20
|
Thurman JM, Frazer-Abel A, Holers VM. The Evolving Landscape for Complement Therapeutics in Rheumatic and Autoimmune Diseases. Arthritis Rheumatol 2017; 69:2102-2113. [PMID: 28732131 PMCID: PMC5659941 DOI: 10.1002/art.40219] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Abstract
The complement system is increasingly understood to play major roles in the pathogenesis of human inflammatory and autoimmune diseases. Because of this situation, there are rapidly expanding commercial efforts to develop novel complement inhibitors and effector pathway-modulating drugs. This review provides insights into the evolving understanding of the complement system components, mechanisms of activation within and across the 3 pathways (classical, alternative, and lectin), how the pathways are normally controlled and then dysregulated in target tissues, and what diseases are known to be, in large part, complement-dependent through the successful development and approval of complement therapeutics in patients. Mechanisms of complement activation in rheumatoid arthritis, lupus, and thrombotic microangiopathies are also illustrated. In addition, the specific therapeutic drugs that are both approved and under development are discussed in the context of both nonrheumatic and rheumatic diseases. Finally, the methods by which the complement system can be assessed in humans through biomarker studies are outlined, with the goal of understanding, in specific patients, how the system is functioning.
Collapse
Affiliation(s)
- Joshua M. Thurman
- University of Colorado Denver, Division of Nephrology and Hypertension, Aurora, CO, USA
| | - Ashley Frazer-Abel
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
- Exsera BioLabs, University of Colorado Denver, Aurora, CO, USA
| | - V. Michael Holers
- University of Colorado Denver, Division of Rheumatology, Aurora, CO, USA
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Systemic sclerosis (SSc) is a heterogeneous autoimmune disease which has defined three hallmarks: Small vessel vasculopathy, production of autoantibodies and fibroblast dysfunction. The exact aetiology of the disease remains unknown, due to the complex nature of the cellular signalling pathways involved. However, there is strong and consistent evidence that the innate system, in particular toll-like receptor signalling, is contributing to the progression and perhaps onset of systemic sclerosis. In light of this evidence, this review examines the role of innate immunity in systemic sclerosis and where appropriate suggests avenues for therapeutic modulation in SSc. RECENT FINDINGS Multiple lines of evidence suggest that Toll-like receptors (TLRs) are dysregulated and emerging evidence suggests that many endogenous ligands are also elevated in the disease leading to 'sterile inflammation' and ultimately the induction of fibrosis. Currently, no effective therapy exists and exploiting the innate immune system perturbation may be one possible avenue. Innate immune dysregulation is key in SSc pathogenesis and may represent a novel target.
Collapse
|
22
|
Ghebrehiwet B, Kaplan AP, Joseph K, Peerschke EIB. The complement and contact activation systems: partnership in pathogenesis beyond angioedema. Immunol Rev 2017; 274:281-289. [PMID: 27782339 DOI: 10.1111/imr.12469] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The blood plasma contains four biologically important proteolytic cascades, which probably evolved from the same ancestral gene. This in part may explain why each cascade has very similar "initiating trigger" followed by sequential and cascade-like downstream enzymatic activation pattern. The four cascades are: the complement system, the blood clotting cascade, the fibrinolytic system, and the kallikrein-kinin system. Although much has been written about the interplay between all these enzymatic cascades, the cross-talk between the complement and the kinin generating systems has become particularly relevant as this interaction results in the generation of nascent molecules that have significant impact in various inflammatory diseases including angioedema and cancer. In this review, we will focus on the consequences of the interplay between the two systems by highlighting the role of a novel molecular link called gC1qR. Although this protein was first identified as a receptor for C1q, it is now recognized as a multiligand binding cellular protein, which serves not only as C1q receptor, but also as high affinity (KD ≤ 0.8 nM) binding site for both high molecular weight kininogen (HK) and factor XII (FXII). At inflammatory sites, where atherogenic factors such as immune complexes and/or pathogens can activate the endothelial cell into a procoagulant and proinflammatory surface, the two pathways are activated to generate vasoactive peptides that contribute in various ways to the inflammatory processes associated with numerous diseases. More importantly, since recent observations strongly suggest an important role for both pathways in cancer, we will focus on how a growing tumor cluster can employ the byproducts derived from the two activation systems to ensure not only its survival and growth, but also its escape into distal sites of colonization.
Collapse
Affiliation(s)
- Berhane Ghebrehiwet
- The Departments of Medicine and Pathology, Stony Brook University, Stony Brook, NY, USA.
| | - Allen P Kaplan
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Kusumam Joseph
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Ellinor I B Peerschke
- The Department of Laboratory Medicine, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, NY, USA.,The Department of Pathology, Weill-Cornell Medical College, New York, NY, USA
| |
Collapse
|
23
|
Holt BA, Bellavia MC, Potter D, White D, Stowell SR, Sulchek T. Fc microparticles can modulate the physical extent and magnitude of complement activity. Biomater Sci 2017; 5:463-474. [PMID: 28067347 PMCID: PMC5330945 DOI: 10.1039/c6bm00608f] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The complement system is an integral component of the humoral immune system, and describes a cascade of interacting proteins responsible for the opsonization and lysis of foreign pathogens, in addition to the recruitment of immune cells. However, complement activation is also implicated in the progression and complication of immune dysfunctions such as sepsis. Microparticle (MP) biomaterials capable of tuning the local magnitude of serum complement activation could improve complement-mediated cytotoxicity to serum-resistant bacteria or calm an overactive immune response during sepsis. We demonstrate that model Fc-functionalized microparticles can be designed to either enhance or diminish the local cytotoxic effect of complement activation in human serum. The particles were formed with either the antibody Fc domains oriented outward from the particle surface or randomly adsorbed in a non-oriented fashion. In the oriented Fc form, complement products were directly sequestered to the particle surface, including C5a, a potent anaphylatoxin that, when elevated, is associated with poor sepsis prognosis. The oriented particle also lowered the cytotoxicity of serum and thus decreased the antibiotic effect when compared to serum alone. Conversely, the non-oriented microparticles were found to sequester similar levels of C5a, but much lower levels of iC3b and TCC on the microparticle surface, thereby increasing the amount of the soluble terminal complement complex. In addition, the non-oriented microparticles extend the distance over which TCC forms and enhance serum cytotoxicity to bacteria. Together, these two types of complement-modulating particles provide the first biomaterial that can functionally modify the range of complement activation at sites distant from the particle surface. Thus, biomaterials that exploit Fc presentation provide new possibilities to functionally modulate complement activation to achieve a desired clinical result.
Collapse
Affiliation(s)
- Brandon Alexander Holt
- Wallace H. Coulter Department of Biomedical Engineering at the Georgia Institute of Technology and Emory University, Atlanta, GA, USA. and The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael C Bellavia
- Wallace H. Coulter Department of Biomedical Engineering at the Georgia Institute of Technology and Emory University, Atlanta, GA, USA. and The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Daniel Potter
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA and The G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - David White
- United States Department of Agriculture, National Centers for Animal Health, Ames, Iowa, USA
| | - Sean R Stowell
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Todd Sulchek
- Wallace H. Coulter Department of Biomedical Engineering at the Georgia Institute of Technology and Emory University, Atlanta, GA, USA. and The G. W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
24
|
Abstract
The complement system, which consists of three independent but interacting pathways, constitutes a powerful arm of innate immunity. Its major function is to recognize and destroy pathogenic microorganisms as well as eliminate modified self-antigens. Although it is a fine-tuned system with innate capacity to discriminate self from non-self as well as danger from non-danger signals, an unwarranted activation can nonetheless occur and cause tissue destruction. To prevent such activation, specific regulators present both in plasma and on the cell surface tightly control it. Data accumulated over the past four decades have also shown that the complement system is capable of not only cross-talk with the activation cascades of plasma––i.e. blood coagulation, contact activation, and the kinin/kallikrein system––but also serving as a bridge between innate and adaptive immunity. It is for these reasons that the various activation steps of the complement system have been recently targeted for therapy to treat diseases in which the role of complement is beyond doubt. This trend will certainly continue for years to come, especially as novel concepts guiding the field into areas never contemplated before are continuing to be discovered.
Collapse
Affiliation(s)
- Berhane Ghebrehiwet
- The Departments of Medicine and Pathology, Stony Brook University School of Medicine, Health Sciences Center, New York, USA
| |
Collapse
|
25
|
Wiese A, Grünewald P, Schaper KJ, Seydel U. Influence of acyl chain fluidity on the lipopolysaccharide-induced activation of complement. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519010070020901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Lipopolysaccharides (LPSs, endotoxins) are the major amphiphilic constituents of the outer leaflet of the outer membrane of Gram-negative bacteria. They are known to activate the complement cascade to form lytic membrane pores. Here, we study the influence of the fluidity of the acyl chains of LPSs and lipid As on the formation of lytic pores. To this end, we have performed electrical measurements on asymmetric planar endotoxin/phospholipid bilayers as a reconstitution model of the outer membrane using two deep rough mutant LPSs (from Escherichia coli strains WBB01 and WBB25) and two lipid As (from E. coli WBB25 and Rhodobacter sphaeroides). The two LPSs and the two lipid As each differ in their acylation pattern which is correlated with the fluidity. The addition of human serum to the endotoxin side of the bilayers led to the formation of membrane pores, and pore formation correlated in each case with acyl chain fluidity, i.e. time required for the first lytic pore to be formed was shorter for the more fluid endotoxin. Furthermore, in the case of LPSs, the activation rate was higher for the more fluid membrane and the respective bacteria had a higher susceptibility to the growth inhibitory action of serum.
Collapse
Affiliation(s)
- Andre Wiese
- Research Center Borstel, Center for Medicine and Biosciences, Division of Biophysics, Borstel, Germany
| | - Petra Grünewald
- Research Center Borstel, Center for Medicine and Biosciences, Division of Biophysics, Borstel, Germany
| | - Klaus-Jürgen Schaper
- Research Center Borstel, Center for Medicine and Biosciences, Division of Biophysics, Borstel, Germany
| | - Ulrich Seydel
- Research Center Borstel, Center for Medicine and Biosciences, Division of Biophysics, Borstel, Germany,
| |
Collapse
|
26
|
Vogel S, Grabski E, Buschjäger D, Klawonn F, Döring M, Wang J, Fletcher E, Bechmann I, Witte T, Durisin M, Schraven B, Mangsbo SM, Schönfeld K, Czeloth N, Kalinke U. Antibody induced CD4 down-modulation of T cells is site-specifically mediated by CD64(+) cells. Sci Rep 2015; 5:18308. [PMID: 26670584 PMCID: PMC4680940 DOI: 10.1038/srep18308] [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: 08/12/2015] [Accepted: 11/16/2015] [Indexed: 02/04/2023] Open
Abstract
Treatment of PBMC with the CD4-specific mAb BT-061 induces CD4 down-modulation of T cells. Here we report that addition of BT-061 to purified T cells did not confer this effect, whereas incubation of T cells in BT-061 coated wells restored CD4 down-modulation. These results implied that Fcγ receptor mediated cell-cell interactions played a role. In consistence with this hypothesis PBMC depleted of CD64+ monocytes did not confer CD4 down-modulation of BT-061 decorated T cells. Strikingly, CD4 down-modulation was observed in BT-061 treated synovial fluid punctuated from patients’ inflamed joints that comprised enhanced numbers of CD64+ cells. In contrast, in a circulating whole blood system injection of BT-061 did not induce CD4 down-modulation, due to CD64 saturation by serum IgG. Similarly, tonsil derived mononuclear cells devoid of CD64+ cells did not show CD4 down-modulation, whereas addition of blood derived monocytes restored the effect. Thus, the interaction of BT-061 decorated T cells with CD64+ cells is needed for CD4 down-modulation, implying that in patients BT-061 would primarily induce CD4 down-modulation at inflammatory sites. These results highlight the need not only to examine the interaction of a given mAb with single FcγR, but also the immunological environment that is appropriate to support such interactions.
Collapse
Affiliation(s)
- Stephanie Vogel
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Feodor-Lynen-Straße 7, D30625 Hannover
| | - Elena Grabski
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Feodor-Lynen-Straße 7, D30625 Hannover
| | - Daniela Buschjäger
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Feodor-Lynen-Straße 7, D30625 Hannover
| | - Frank Klawonn
- Department of Computer Science, Ostfalia University of Applied Sciences, Salzdahlumer Straße 46/48, D38302 Wolfenbüttel, Germany.,Biostatistics, Helmholtz Centre for Infection Research, Inhoffenstraße 7, D38124 Braunschweig, Germany
| | - Marius Döring
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Feodor-Lynen-Straße 7, D30625 Hannover
| | - Junxi Wang
- Biostatistics, Helmholtz Centre for Infection Research, Inhoffenstraße 7, D38124 Braunschweig, Germany
| | - Erika Fletcher
- Department of Immunology Genetics and Pathology, Uppsala University, Rudbeck Laboratory, S75185 Uppsala, Sweden.,Immuneed AB, S-756 52, Uppsala, Sweden
| | - Ingo Bechmann
- Institute for Anatomy, University Leipzig, Liebigstraße 13, D04103 Leipzig
| | - Torsten Witte
- Clinic for Immunology and Rheumatology, Hannover Medical School, Carl-Neuberg-Straße 1, D30625 Hannover, Germany
| | - Martin Durisin
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Straße 1, D30625 Hannover, Germany
| | - Burkhart Schraven
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, D39120 Magdeburg, Germany.,Department of Immune Control, Helmholtz Centre for Infection Research, Inhoffenstrß2 7, D38124 Braunschweig, Germany
| | - Sara M Mangsbo
- Department of Immunology Genetics and Pathology, Uppsala University, Rudbeck Laboratory, S75185 Uppsala, Sweden.,Immuneed AB, S-756 52, Uppsala, Sweden
| | - Kurt Schönfeld
- Biotest AG, Dreieich, Landsteinerstrasse 5, D63303 Dreieich, Germany
| | - Niklas Czeloth
- Biotest AG, Dreieich, Landsteinerstrasse 5, D63303 Dreieich, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Feodor-Lynen-Straße 7, D30625 Hannover
| |
Collapse
|
27
|
Abstract
Throughout evolution, organisms have developed means to contain wounds by simultaneously limiting bleeding and eliminating pathogens and damaged host cells via the recruitment of innate defense mechanisms. Disease emerges when there is unchecked activation of innate immune and/or coagulation responses. A key component of innate immunity is the complement system. Concurrent excess activation of coagulation and complement - two major blood-borne proteolytic pathways - is evident in numerous diseases, including atherosclerosis, diabetes, venous thromboembolic disease, thrombotic microangiopathies, arthritis, cancer, and infectious diseases. Delineating the cross-talk between these two cascades will uncover novel therapeutic insights.
Collapse
Affiliation(s)
- E M Conway
- Centre for Blood Research, Life Sciences Institute, Division of Hematology, Department of Medicine, Faculty of Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
| |
Collapse
|
28
|
Abstract
Although new activation and regulatory mechanisms are still being identified, the basic architecture of the complement system has been known for decades. Two major roles of complement are to control certain bacterial infections and to promote clearance of apoptotic cells. In addition, although inappropriate complement activation has long been proposed to cause tissue damage in human inflammatory and autoimmune diseases, whether this is indeed true has been uncertain. However, recent studies in humans, especially those using newly available biological therapeutics, have now clearly demonstrated the pathophysiologic importance of the complement system in several rare diseases. Beyond these conditions, recent genetic studies have strongly supported an injurious role for complement in a wide array of human inflammatory, degenerative, and autoimmune diseases. This review includes an overview of complement activation, regulatory, and effector mechanisms. It then focuses on new understandings gained from genetic studies, ex vivo analyses, therapeutic trials, and animal models as well as on new research opportunities.
Collapse
Affiliation(s)
- V Michael Holers
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045;
| |
Collapse
|
29
|
Giles BM, Boackle SA. Linking complement and anti-dsDNA antibodies in the pathogenesis of systemic lupus erythematosus. Immunol Res 2013; 55:10-21. [PMID: 22941560 DOI: 10.1007/s12026-012-8345-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Systemic lupus erythematosus is a severe autoimmune disease that affects multiple organ systems resulting in diverse symptoms and outcomes. It is characterized by antibody production to a variety of self-antigens, but it is specifically associated with those against anti-dsDNA. Anti-dsDNA antibodies are present before the onset of clinical disease and are associated with severe manifestations of lupus such as glomerulonephritis. Their levels fluctuate with changes in disease activity and, in combination with the levels of complement proteins C3 and C4, are strong indicators of disease flare and treatment response in patients with lupus. The decreased complement levels that are noted during flares of lupus activity are believed to be secondary to increased autoantibody production and immune complex formation that results in tissue damage; however, recent data suggest that complement activation can also drive development of these pathogenic autoantibodies. This review will explore the various roles of complement in the development and pathogenesis of anti-dsDNA antibodies.
Collapse
Affiliation(s)
- Brendan M Giles
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | |
Collapse
|
30
|
DeZern AE, Dorr D, Brodsky RA. Predictors of hemoglobin response to eculizumab therapy in paroxysmal nocturnal hemoglobinuria. Eur J Haematol 2013; 90:16-24. [PMID: 23046169 PMCID: PMC3673783 DOI: 10.1111/ejh.12021] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND Paroxysmal nocturnal hemoglobinuria (PNH) is a clonal, hematopoietic stem cell disorder that manifests with hemolytic anemia and bone marrow failure. Eculizumab has been shown to improve anemia, decrease intravascular hemolysis, and reduce the risk of thrombosis. DESIGN AND METHODS This is a retrospective, single-center study of patients treated with eculizumab and categorized according to response criteria. Complete response (CR) was defined as transfusion independence with normal hemoglobin for age/sex, absence of symptoms, and lactate dehydrogenase <1.5 times the upper limit of normal. A good partial response (GPR) was defined as decreased transfusions from pretreatment and lactate dehydrogenase <1.5 upper limit of normal without thrombosis. These patients did not achieve normal hemoglobins for age and sex. A suboptimal response was defined as unchanged transfusion needs and persistent of symptoms. RESULTS Thirty patients with PNH clones were treated with eculizumab and classified as complete responders (four patients), good partial responders (16), and suboptimal responders (10) over 863 patient-months of treatment. Complete responders had a decrease in red cell clone size, while good partial responders had an increase. Thirteen patients treated did not meet inclusion criteria for the clinical trials of eculizumab due to lack of transfusions or thrombocytopenia; eight had at least a GPR. CONCLUSIONS Eculizumab is efficacious in patients with PNH, but responses can vary and may depend on underlying marrow failure, underlying inflammatory conditions and red cell clone size following treatment. Normalization of hemoglobin with decrease in red cell clone size may predict CR.
Collapse
Affiliation(s)
- Amy E DeZern
- Division of Hematology, Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
31
|
CR2-mediated targeting of complement inhibitors: bench-to-bedside using a novel strategy for site-specific complement modulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:137-54. [PMID: 23402024 DOI: 10.1007/978-1-4614-4118-2_9] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent approval of the first human complement pathway-directed therapeutics, along with high-profile genetic association studies, has catalyzed renewed biopharmaceutical interest in developing drugs that modulate the complement system. Substantial challenges remain, however, that must be overcome before widespread application of complement inhibitors in inflammatory and autoimmune diseases becomes possible. Among these challenges are the following: (1) defining the complement pathways and effector mechanisms that cause tissue injury in humans and determining whether the relative importance of each varies by disease, (2) blocking or modulating, using traditional small molecule or biologic approaches, the function of complement proteins whose circulating levels are very high and whose turnover rates are relatively rapid, especially in the setting of acute and chronic autoimmune diseases, and (3) avoiding infectious complications or impairment of other important physiological functions of complement when using systemically active complement-blocking agents. This chapter will review data that address these challenges to therapeutic development, with a focus on the development of a novel strategy of blocking specific complement pathways by targeting inhibitors using a recombinant portion of the human complement receptor type 2 (CR2/CD21) which specifically targets to sites of local complement C3 activation where C3 fragments are covalently fixed. Recently, the first of these CR2-targeted proteins has entered human phase I studies in the human disease paroxysmal nocturnal hemoglobinuria. The results of murine translational studies using CR2-targeted inhibitors strongly suggest that a guiding principle going forward in complement therapeutic development may well be to focus on developing strategies to modulate the pathway as precisely as possible by physically localizing therapeutic inhibitory effects.
Collapse
|
32
|
Effect of complement on HIV-2 plasma antiviral activity is intratype specific and potent. J Virol 2012; 87:273-81. [PMID: 23077299 DOI: 10.1128/jvi.01640-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus type 2 (HIV-2)-infected individuals develop immunodeficiency with a considerable delay and transmit the virus at rates lower than HIV-1-infected persons. Conceivably, comparative studies on the immune responsiveness of HIV-1- and HIV-2-infected hosts may help to explain the differences in pathogenesis and transmission between the two types of infection. Previous studies have shown that the neutralizing antibody response is more potent and broader in HIV-2 than in HIV-1 infection. In the present study, we have examined further the function of the humoral immune response and studied the effect of complement on the antiviral activity of plasma from singly HIV-1- or HIV-2-infected individuals, as well as HIV-1/HIV-2 dually infected individuals. The neutralization and antibody-dependent complement-mediated inactivation of HIV-1 and HIV-2 isolates were tested in a plaque reduction assay using U87.CD4.CCR5 cells. The results showed that the addition of complement increased intratype antiviral activities of both HIV-1 and HIV-2 plasma samples, although the complement effect was more pronounced with HIV-2 than HIV-1 plasma. Using an area-under-the-curve (AUC)-based readout, multivariate statistical analysis confirmed that the type of HIV infection was independently associated with the magnitude of the complement effect. The analyses carried out with purified IgG indicated that the complement effect was largely exerted through the classical complement pathway involving IgG in both HIV-1 and HIV-2 infections. In summary, these findings suggest that antibody binding to HIV-2 structures facilitates the efficient use of complement and thereby may be one factor contributing to a strong antiviral activity present in HIV-2 infection.
Collapse
|
33
|
Pechtl IC, Neely RK, Dryden DTF, Jones AC, Barlow PN. Use of time-resolved FRET to validate crystal structure of complement regulatory complex between C3b and factor H (N terminus). Protein Sci 2012; 20:2102-12. [PMID: 21936007 DOI: 10.1002/pro.738] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Structural knowledge of interactions amongst the ~ 40 proteins of the human complement system, which is central to immune surveillance and homeostasis, is expanding due primarily to X-ray diffraction of co-crystallized proteins. Orthogonal evidence, in solution, for the physiological relevance of such co-crystal structures is valuable since intermolecular affinities are generally weak-to-medium and inter-domain mobility may be important. In this current work, Förster resonance energy transfer (FRET) was used to investigate the 10 μM K(D) (210 kD) complex between the N-terminal region of the soluble complement regulator, factor H (FH1-4), and the key activation-specific complement fragment, C3b. Using site-directed mutagenesis, seven cysteines were introduced individually at potentially informative positions within the four CCP modules comprising FH1-4, then used for fluorophore attachment. C3b possesses a thioester domain featuring an internal cycloglutamyl cysteine thioester; upon hydrolysis this yields a free thiol (Cys988) that was also fluorescently tagged. Labeled proteins were functionally active as cofactors for cleavage of C3b to iC3b except for FH1-4(Q40C) where conjugation with the fluorophore likely abrogated interaction with the protease, factor I. Time-resolved FRET measurements were undertaken to explore interactions between FH1-4 and C3b in fluid phase and under near-physiological conditions. These experiments confirmed that, as in the cocrystal structure, FH1-4 binds to C3b with CCP module 1 furthest from, and CCP module 4 closest to, the thioester domain, placing subsequent modules of FH near to any surface to which C3b is attached. The data do not rule out flexibility of the thioester domain relative to the remainder of the complex.
Collapse
Affiliation(s)
- Isabell C Pechtl
- EaStCHEM School of Chemistry and Collaborative Optical Spectroscopy, Micromanipulation and Imaging Centre (COSMIC), University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | |
Collapse
|
34
|
Identification of quantitative trait loci in experimental epidermolysis bullosa acquisita. J Invest Dermatol 2012; 132:1409-15. [PMID: 22297639 DOI: 10.1038/jid.2011.466] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epidermolysis bullosa acquisita (EBA) is a chronic mucocutaneous autoimmune skin blistering disease. Several lines of evidence underscore the contribution of autoantibodies against type VII collagen (COL7) to the pathogenesis of EBA. Furthermore, EBA susceptibility is associated with the MHC haplotype in patients (HLA-DR2) and in immunization-induced EBA in mice (H2s). The latter study indicated an additional contribution of non-MHC genes to disease susceptibility. To identify non-MHC genes controlling EBA susceptibility, we intercrossed EBA-susceptible MRL/MpJ with EBA-resistant NZM2410/J and BXD2/TyJ as well as Cast mice. Mice of the fourth generation of this four-way autoimmune-prone advanced intercross line were immunized with a fragment of murine COL7 to induce EBA. Anti-COL7 autoantibodies were detected in 84% of mice, whereas deposition of complement at the dermal-epidermal junction (DEJ) was observed in 50% of the animals; 33% of immunized mice presented with overt clinical EBA. Onset of clinical disease was associated with several quantitative trait loci (QTLs) located on chromosomes 9, 12, 14, and 19, whereas maximum disease severity was linked to QTLs on chromosomes 1, 15, and 19. This more detailed insight into the pathogenesis of EBA may eventually lead to new treatment strategies for EBA and other autoantibody-mediated diseases.
Collapse
|
35
|
Cross-talk between the complement and the kinin system in vascular permeability. Immunol Lett 2011; 140:7-13. [PMID: 21762728 DOI: 10.1016/j.imlet.2011.06.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 06/08/2011] [Accepted: 06/23/2011] [Indexed: 02/01/2023]
Abstract
The endothelium is a continuous physical barrier that regulates coagulation and selective passage of soluble molecules and circulating cells through the vessel wall into the tissue. Due to its anatomic localization, the endothelium may establish contact with components of the complement, the kinin and the coagulation systems which are the main, though not exclusive, inducers of vascular leakage. Although the complement and the kinin systems may act independently, increasing evidence suggest that there is a crosstalk that involve different components of both systems. Activation is required for the function of the two systems which are involved in pathological conditions such as hereditary and acquired angioedema (AE) and vasculitidis. The aim of this review is to discuss the contribution of complement and kinin systems to vascular leakage and the cross-talk between the two systems in the development of AE. This clinical condition is characterized by episodic and recurrent local edema of subcutaneous and submucosal tissues and is due to inherited or acquired C1-INH deficiency. Although the pathogenesis of the swelling in patients with AE was originally thought to be mediated by C2, ample evidence indicate bradykinin (BK) as the most effective mediator even though the possibility that both the complement and the kinin-forming systems may contribute to the edema has not been completely excluded. BK induces endothelial leakage interacting with B2 receptors but other molecules may be involved in the onset and maintenance of AE. In this review we shall discuss the role of B1 receptors and gC1qR/p33 in addition to that of B2 receptors in the onset of AE attacks and the importance of these receptors as new possible molecular targets for therapy.
Collapse
|
36
|
Common polymorphisms in C3, factor B, and factor H collaborate to determine systemic complement activity and disease risk. Proc Natl Acad Sci U S A 2011; 108:8761-6. [PMID: 21555552 DOI: 10.1073/pnas.1019338108] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Common polymorphisms in complement alternative pathway (AP) proteins C3 (C3(R102G)), factor B (fB(R32Q)), and factor H (fH(V62I)) are associated with age-related macular degeneration (AMD) and other pathologies. Our published work showed that fB(R32Q) influences C3 convertase formation, whereas fH(V62I) affects factor I cofactor activity. Here we show how C3(R102G) (C3S/F) influences AP activity. In hemolysis assays, C3(102G) activated AP more efficiently (EC(50) C3(102G): 157 nM; C3(102R): 191 nM; P < 0.0001). fB binding kinetics and convertase stability were identical, but native and recombinant fH bound more strongly to C3b(102R) (K(D) C3b(102R): 1.0 μM; C3b(102G): 1.4 μM; P < 0.0001). Accelerated decay was unaltered, but fH cofactor activity was reduced for C3b(102G), favoring AP amplification. Combining disease "risk" variants (C3(102G), fB(32R), and fH(62V)) in add-back assays yielded sixfold higher hemolytic activity compared with "protective" variants (C3(102R), fB(32Q), and fH(62I); P < 0.0001). These data introduce the concept of a functional complotype (combination of polymorphisms) defining complement activity in an individual, thereby influencing susceptibility to AP-driven disease.
Collapse
|
37
|
Holers VM. The complement system in systemic lupus erythematosus. Rheumatology (Oxford) 2011. [DOI: 10.1016/b978-0-323-06551-1.00020-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
38
|
Banda NK, Takahashi M, Levitt B, Glogowska M, Nicholas J, Takahashi K, Stahl GL, Fujita T, Arend WP, Holers VM. Essential role of complement mannose-binding lectin-associated serine proteases-1/3 in the murine collagen antibody-induced model of inflammatory arthritis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:5598-606. [PMID: 20870940 PMCID: PMC3157645 DOI: 10.4049/jimmunol.1001564] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gene-targeted mice deficient in the complement mannose-binding lectin-associated serine protease-1 and -3 (MASP1/3(-/-)) express only the zymogen of factor D (pro-factor D [pro-Df]), a necessary component of the alternative pathway (AP). We used the murine collagen Ab-induced arthritis (CAIA) model, in which the AP is unique among complement pathways in being both necessary and sufficient for disease induction, to determine whether MASP-1/3 are required in vivo for the development of tissue injury. Disease activity scores, complement C3 tissue deposition in the joint, and histopathologic injury scores were markedly decreased in MASP1/3(-/-) as compared with wild-type (WT) mice. MASP-1 protein was immunochemically localized to synovial cells of knees of WT mice with arthritis. Pro-Df was present in both synovial cells and chondrocytes of knees of WT and MASP1/3(-/-) mice without arthritis, with increased amounts present in synovial cells of WT mice with CAIA. No conversion of pro-Df to mature Df was detectable in the serum of MASP1/3(-/-) mice during the evolution of CAIA. C3 activation and deposition as well as C5a generation induced in vitro by adherent anti-type II collagen mAbs were absent using sera from MASP1/3(-/-) mice under conditions in which only the AP was active. The addition of human Df fully reconstituted in vitro C3 activation and C5a generation using sera from MASP1/3(-/-) mice. Our studies demonstrate for the first time, to our knowledge, the absolute requirement for the activity of MASP-1 protein in autoimmune-associated inflammatory tissue injury in vivo through activation of the AP of complement by cleavage of pro-Df to mature Df.
Collapse
Affiliation(s)
- Nirmal K. Banda
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045
| | - Minoru Takahashi
- Department of Immunology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Brandt Levitt
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045
| | - Magdalena Glogowska
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045
| | - Jessica Nicholas
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045
| | - Kazue Takahashi
- Developmental Immunology, Massachusetts General Hospital for Children, Boston, MA 02114
| | - Gregory L. Stahl
- Center of Experimental Therapeutics and Reperfusion Injury, Brigham and Women’s Hospital, Boston, MA 02115
| | - Teizo Fujita
- Department of Immunology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - William P. Arend
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045
| | - V. Michael Holers
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Immunology, University of Colorado School of Medicine, Aurora, CO 80045
| |
Collapse
|
39
|
Leung VWY, Yun S, Botto M, Mason JC, Malik TH, Song W, Paixao-Cavalcante D, Pickering MC, Boyle JJ, Haskard DO. Decay-accelerating factor suppresses complement C3 activation and retards atherosclerosis in low-density lipoprotein receptor-deficient mice. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1757-67. [PMID: 19729477 DOI: 10.2353/ajpath.2009.090183] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Decay-accelerating factor (DAF; CD55) is a membrane protein that regulates complement pathway activity at the level of C3. To test the hypothesis that DAF plays an essential role in limiting complement activation in the arterial wall and protecting from atherosclerosis, we crossed DAF gene targeted mice (daf-1(-/-)) with low-density lipoprotein-receptor deficient mice (Ldlr(-/-)). Daf-1(-/-)Ldlr(-/-) mice had more extensive en face Sudan IV staining of the thoracoabdominal aorta than Ldlr(-/-) mice, both following a 12-week period of low-fat diet or a high-fat diet. Aortic root lesions in daf-1(-/-)Ldlr(-/-) mice on a low-fat diet showed increased size and complexity. DAF deficiency increased deposition of C3d and C5b-9, indicating the importance of DAF for downstream complement regulation in the arterial wall. The acceleration of lesion development in the absence of DAF provides confirmation of the proinflammatory and proatherosclerotic potential of complement activation in the Ldlr(-/-) mouse model. Because upstream complement activation is potentially protective, this study underlines the importance of DAF in shielding the arterial wall from the atherogenic effects of complement.
Collapse
Affiliation(s)
- Viola W Y Leung
- Vascular Science Section, Imperial College, National Heart and Lung Institute, the Division of Investigative Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Complement inhibiting surface proteins of pathogenic bacteria provide candidates for vaccines because of two reasons. First, an immune response against them would recognize the microbes and secondly, it would neutralize the key bacterial virulence mechanism. Prerequisites for a vaccine protein include the following: (i) it should show limited variability, (ii) it should be immunogenic and the immune response against it should cover a sufficiently broad range of microbial strains, (iii) it should not be hidden beneath a capsule, long LPS O-polysaccharide side chains or a protein coat and (iv) it should not raise unwanted immune responses against host structures. Bacterial complement inhibitors often act by binding the soluble inhibitors factor H or C4 bp, by blocking C3 or C5 activation or by enzymatically cleaving key complement components. Inhibitors have been found from all major types of pathogens and may offer promise as rational vaccine candidates for preventing diseases such as meningococcal meningitis, systemic pneumococcal or group B streptococcal disease and Lyme borreliosis.
Collapse
|
41
|
Fraiture M, Baxter RHG, Steinert S, Chelliah Y, Frolet C, Quispe-Tintaya W, Hoffmann JA, Blandin SA, Levashina EA. Two mosquito LRR proteins function as complement control factors in the TEP1-mediated killing of Plasmodium. Cell Host Microbe 2009; 5:273-84. [PMID: 19286136 DOI: 10.1016/j.chom.2009.01.005] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 10/31/2008] [Accepted: 01/17/2009] [Indexed: 10/21/2022]
Abstract
Plasmodium development within Anopheles mosquitoes is a vulnerable step in the parasite transmission cycle, and targeting this step represents a promising strategy for malaria control. The thioester-containing complement-like protein TEP1 and two leucine-rich repeat (LRR) proteins, LRIM1 and APL1, have been identified as major mosquito factors that regulate parasite loads. Here, we show that LRIM1 and APL1 are required for binding of TEP1 to parasites. RNAi silencing of the LRR-encoding genes results in deposition of TEP1 on Anopheles tissues, thereby depleting TEP1 from circulation in the hemolymph and impeding its binding to Plasmodium. LRIM1 and APL1 not only stabilize circulating TEP1, they also stabilize each other prior to their interaction with TEP1. Our results indicate that three major antiparasitic factors in mosquitoes jointly function as a complement-like system in parasite killing, and they reveal a role for LRR proteins as complement control factors.
Collapse
Affiliation(s)
- Malou Fraiture
- UPR 9022 CNRS, AVENIR group Inserm, Institut de Biologie Moléculaire et Cellulaire, 15 rue René Descartes, 67084 Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Functional basis of protection against age-related macular degeneration conferred by a common polymorphism in complement factor B. Proc Natl Acad Sci U S A 2009; 106:4366-71. [PMID: 19255449 DOI: 10.1073/pnas.0812584106] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mutations and polymorphisms in complement genes have been linked with numerous rare and prevalent disorders, implicating dysregulation of complement in pathogenesis. The 3 common alleles of factor B (fB) encode Arg (fB(32R)), Gln (fB(32Q)), or Trp (fB(32W)) at position 32 in the Ba domain. The fB(32Q) allele is protective for age-related macular degeneration, the commonest cause of blindness in developed countries. Factor B variants were purified from plasma of homozygous individuals and were tested in hemolysis assays. The protective variant fB(32Q) had decreased activity compared with fB(32R). Biacore comparison revealed markedly different proenzyme formation; fB(32R) bound C3b with 4-fold higher affinity, and formation of activated convertase was enhanced. Binding and functional differences were confirmed with recombinant fB(32R) and fB(32Q); an intermediate affinity was revealed for fB(32W). To confirm contribution of Ba to binding, affinity of Ba for C3b was determined. Ba-fB(32R) had 3-fold higher affinity compared with Ba-fB(32Q). We demonstrate that the disease-protective effect of fB(32Q) is consequent on decreased potential to form convertase and amplify complement activation. Knowledge of the functional consequences of polymorphisms in complement activators and regulators will aid disease prediction and inform targeting of diagnostics and therapeutics.
Collapse
|
43
|
Abstract
OBJECTIVE To examine complement cascade activation after an acute psychological stress task. Psychological stress has been implicated in the exacerbation of inflammatory disorders. Although the complement cascade is a key component of these inflammatory processes, there has been little research regarding its susceptibility to stress. METHODS In experiment 1, 38 healthy participants completed an 8-minute psychological stress task. Complement components were assessed from blood samples taken by venipuncture, at rest and immediately post task. In experiment 2, 40 participants undertook a similar task; blood samples were collected from a cannula at rest, immediately post task, and after 30 and 60 minutes of recovery. In experiment 3, 40 participants were exposed to both a stress and a control session. Session order was counterbalanced and, on both occasions, we received blood samples from half the participants via a cannula and the other half by repeated venipuncture. RESULTS In experiment 1, C3a levels increased significantly from rest to task, indicating complement cascade activation. In experiment 2, we found that both C3a and Factor Bb increased significantly from rest to task and recovered by 30 and 60 minutes. C5a rose significantly 30 minutes after completion of the stress task. In experiment 3, C3a increased in response to the mental stress task, whereas it decreased slightly during the control session. There was no significant effect of blood taking method. CONCLUSIONS These experiments demonstrate that the complement cascade is susceptible to acute psychological stress and suggest a potential mechanism for stress-induced inflammatory activation in individuals with inflammatory disorders.
Collapse
|
44
|
Yuhki KI, Ushikubi F, Naraba H, Ueno A, Kato H, Kojima F, Narumiya S, Sugimoto Y, Matsushita M, Oh-ishi S. Prostaglandin I2 Plays a Key Role in Zymosan-Induced Mouse Pleurisy. J Pharmacol Exp Ther 2008; 325:601-9. [DOI: 10.1124/jpet.107.134494] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
45
|
Banda NK, Takahashi K, Wood AK, Holers VM, Arend WP. Pathogenic complement activation in collagen antibody-induced arthritis in mice requires amplification by the alternative pathway. THE JOURNAL OF IMMUNOLOGY 2007; 179:4101-9. [PMID: 17785849 DOI: 10.4049/jimmunol.179.6.4101] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immune complex-induced inflammation can be mediated by the classical pathway of complement. However, using mice genetically deficient in factor B or C4, we have shown that the collagen Ab-induced model of arthritis requires the alternative pathway of complement and is not dependent on the classical pathway. We now demonstrate that collagen Ab-induced arthritis is not altered in mice genetically deficient in either C1q or mannose-binding lectins A and C, or in both C1q and mannose-binding lectins. These in vivo results prove the ability of the alternative pathway to carry out pathologic complement activation in the combined absence of intact classical and lectin pathways. C3 activation was also examined in vitro by adherent collagen-anti-collagen immune complexes using sera from normal or complement-deficient mice. These results confirm the ability of the alternative pathway to mediate immune complex-induced C3 activation when C4 or C1q, or both C1q and mannose-binding lectins, are absent. However, when all three activation pathways of complement are intact, initiation by immune complexes occurs primarily by the classical pathway. These results indicate that the alternative pathway amplification loop, with its ability to greatly enhance C3 activation, is necessary to mediate inflammatory arthritis induced by adherent immune complexes.
Collapse
Affiliation(s)
- Nirmal K Banda
- Division of Rheumatology B115, University of Colorado at Denver and Health Sciences Center, Aurora, CO 80045, USA
| | | | | | | | | |
Collapse
|
46
|
Spitzer D, Mitchell LM, Atkinson JP, Hourcade DE. Properdin can initiate complement activation by binding specific target surfaces and providing a platform for de novo convertase assembly. THE JOURNAL OF IMMUNOLOGY 2007; 179:2600-8. [PMID: 17675523 DOI: 10.4049/jimmunol.179.4.2600] [Citation(s) in RCA: 228] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Complement promotes the rapid recognition and elimination of pathogens, infected cells, and immune complexes. The biochemical basis for its target specificity is incompletely understood. In this report, we demonstrate that properdin can directly bind to microbial targets and provide a platform for the in situ assembly and function of the alternative pathway C3 convertases. This mechanism differs from the standard model wherein nascent C3b generated in the fluid phase attaches nonspecifically to its targets. Properdin-directed complement activation occurred on yeast cell walls (zymosan) and Neisseria gonorrhoeae. Properdin did not bind wild-type Escherichia coli, but it readily bound E. coli LPS mutants, and the properdin-binding capacity of each strain correlated with its respective serum-dependent AP activation rate. Moreover, properdin:single-chain Ab constructs were used to direct serum-dependent complement activation to novel targets. We conclude properdin participates in two distinct complement activation pathways: one that occurs by the standard model and one that proceeds by the properdin-directed model. The properdin-directed model is consistent with a proposal made by Pillemer and his colleagues >50 years ago.
Collapse
Affiliation(s)
- Dirk Spitzer
- Division of Rheumatology, Department of Medicine, School of Medicine, Washington University, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
47
|
Onda K, Ohi H, Tamano M, Ohsawa I, Wakabayashi M, Horikoshi S, Fujita T, Tomino Y. Hypercomplementemia in adult patients with IgA nephropathy. J Clin Lab Anal 2007; 21:77-84. [PMID: 17385664 PMCID: PMC6649110 DOI: 10.1002/jcla.20154] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
IgA nephropathy (IgAN) is the most common form of chronic glomerulonephritis. Although glomerular deposition of complement components is well known, the evidence of serological complement activation in IgAN is inconclusive. We hypothesized that serum levels of complement components and regulatory proteins in patients with IgAN are correlated with its pathogenesis. In the present study we measured complement components in 50 patients with IgAN and 50 healthy volunteers. C5, C1 inhibitor, factor B, C4 binding protein, factor H, and factor I were measured with the use of single radial immunodiffusion. Mannose-binding lectin (MBL) and properdin (P) were measured by enzyme-linked immunosorbent assay (ELISA). The correlations among complements in the sera of patients with clinical gradings for IgAN (i.e., the good prognosis group, relatively good prognosis group, relatively poor prognosis group, and poor prognosis group) were evaluated. CH50, C4, factor B, P, factor I, and factor H were significantly higher in IgAN patients than in healthy controls. There were significant correlations between C5 and C4 binding protein, between C3 and C5, or between C4 and factor B in patients with IgAN. In the poor prognosis group, C4 binding protein was significantly higher than in the other groups of IgAN patients. hypercomplementemia occurs in IgAN and is associated with an increase in complement regulatory protein (CRP). C4 binding protein analyses can be used to predict disease prognosis.
Collapse
Affiliation(s)
- Kisara Onda
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Juntendo University, Tokyo, Japan
| | - Hiroyuki Ohi
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Juntendo University, Tokyo, Japan
| | - Mariko Tamano
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Juntendo University, Tokyo, Japan
| | - Isao Ohsawa
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Juntendo University, Tokyo, Japan
| | - Michiro Wakabayashi
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Juntendo University, Tokyo, Japan
| | - Satoshi Horikoshi
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Juntendo University, Tokyo, Japan
| | - Teizo Fujita
- Department of Immunology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yasuhiko Tomino
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
48
|
Mihai S, Chiriac MT, Takahashi K, Thurman JM, Holers VM, Zillikens D, Botto M, Sitaru C. The Alternative Pathway of Complement Activation Is Critical for Blister Induction in Experimental Epidermolysis Bullosa Acquisita. THE JOURNAL OF IMMUNOLOGY 2007; 178:6514-21. [PMID: 17475881 DOI: 10.4049/jimmunol.178.10.6514] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Epidermolysis bullosa acquisita is a subepidermal blistering disease associated with tissue-bound and circulating autoantibodies against type VII collagen, a major constituent of the dermal-epidermal junction. The passive transfer of Abs against type VII collagen into mice induces a subepidermal blistering disease dependent upon activation of terminal complement components. To further dissect the role of the different complement activation pathways in this model, we injected C1q-deficient, mannan-binding lectin-deficient, and factor B-deficient mice with rabbit Abs against murine type VII collagen. The development and evolution of blistering had a similar pattern in mannan-binding lectin-deficient and control mice and was initially only marginally less extensive in C1q-deficient mice compared with controls. Importantly, factor B-deficient mice developed a delayed and significantly less severe blistering disease compared with factor B-sufficient mice. A significantly lower neutrophilic infiltration was observed in factor B-deficient mice compared with controls and local reconstitution with granulocytes restored the blistering disease in factor B-deficient mice. Our study provides the first direct evidence for the involvement of the alternative pathway in an autoantibody-induced blistering disease and should facilitate the development of new therapeutic strategies for epidermolysis bullosa acquisita and related autoimmune diseases.
Collapse
Affiliation(s)
- Sidonia Mihai
- Department of Dermatology, University of Lübeck, Lübeck, Germany, and Department of Pediatrics, Laboratory of Developmental Immunology, Massachusetts General Hospital, Boston 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Harris CL, Pettigrew DM, Lea SM, Morgan BP. Decay-accelerating factor must bind both components of the complement alternative pathway C3 convertase to mediate efficient decay. THE JOURNAL OF IMMUNOLOGY 2007; 178:352-9. [PMID: 17182573 DOI: 10.4049/jimmunol.178.1.352] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Decay-accelerating factor (DAF; CD55) inhibits the complement (C) cascade by dissociating the multimolecular C3 convertase enzymes central to amplification. We have previously demonstrated using surface plasmon resonance (Biacore International) that DAF mediates decay of the alternative pathway C3 convertase, C3bBb, but not of the inactive proenzyme, C3bB, and have shown that the major site of interaction is with the larger cleavage subunit factor B (Bb) subunit. In this study, we dissect these interactions and demonstrate that the second short consensus repeat (SCR) domain of DAF (SCR2) interacts only with Bb, whereas SCR4 interacts with C3b. Despite earlier studies that found SCR3 to be critical to DAF activity, we find that SCR3 does not directly interact with either subunit. Furthermore, we demonstrate that properdin, a positive regulator of the alternative pathway, does not directly interact with DAF. Extending from studies of binding to decay-accelerating activity, we show that truncated forms of DAF consisting of SCRs 2 and 3 bind the convertase stably via SCR2-Bb interactions but have little functional activity. In contrast, an SCR34 construct mediates decay acceleration, presumably due to SCR4-C3b interactions demonstrated above, because SCR3 alone has no binding or functional effect. We propose that DAF interacts with C3bBb through major sites in SCR2 and SCR4. Binding to Bb via SCR2 increases avidity of binding, concentrating DAF on the active convertase, whereas more transient interactions through SCR4 with C3b directly mediate decay acceleration. These data provide new insights into the mechanisms involved in C3 convertase decay by DAF.
Collapse
Affiliation(s)
- Claire L Harris
- Department of Medical Biochemistry and Immunology, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | | | | | |
Collapse
|
50
|
Kraus DM, Elliott GS, Chute H, Horan T, Pfenninger KH, Sanford SD, Foster S, Scully S, Welcher AA, Holers VM. CSMD1 is a novel multiple domain complement-regulatory protein highly expressed in the central nervous system and epithelial tissues. THE JOURNAL OF IMMUNOLOGY 2006; 176:4419-30. [PMID: 16547280 DOI: 10.4049/jimmunol.176.7.4419] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In this study, we describe the identification and in vitro functional activity of a novel multiple domain complement regulatory protein discovered based on its homology to short consensus repeat (SCR)-containing proteins of the regulators of complement activation (RCA) gene family. The rat cDNA encodes a predicted 388-kDa protein consisting of 14 N-terminal CUB domains that are separated from each other by a SCR followed by 15 tandem SCR domains, a transmembrane domain, and a short cytoplasmic tail. This protein is the homolog of the human protein of unknown function called the CUB and sushi multiple domains 1 (CSMD1) protein. A cloning strategy that incorporates the two C-terminal CUB-SCR domains and 12 of the tandem SCR repeats was used to produce a soluble rat CSMD1 protein. This protein blocked classical complement pathway activation in a comparable fashion with rat Crry but did not block alternative pathway activation. Analysis of CSMD1 mRNA expression by in situ hybridization and immunolabeling of neurons indicates that the primary sites of synthesis are the developing CNS and epithelial tissues. Of particular significance is the enrichment of CSMD1 in the nerve growth cone, the amoeboid-leading edge of the growing neuron. These results suggest that CSMD1 may be an important regulator of complement activation and inflammation in the developing CNS, and that it may also play a role in the context of growth cone function.
Collapse
Affiliation(s)
- Damian M Kraus
- Division of Rheumatology, University of Colorado Health Sciences Center, Denver, 80262, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|