1
|
Xu Z, Xu B, Lundström SL, Moreno-Giró À, Zhao D, Martin M, Lönnblom E, Li Q, Krämer A, Ge C, Cheng L, Liang B, Tong D, Stawikowska R, Blom AM, Fields GB, Zubarev RA, Holmdahl R. A subset of type-II collagen-binding antibodies prevents experimental arthritis by inhibiting FCGR3 signaling in neutrophils. Nat Commun 2023; 14:5949. [PMID: 37741824 PMCID: PMC10517938 DOI: 10.1038/s41467-023-41561-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/06/2023] [Indexed: 09/25/2023] Open
Abstract
Rheumatoid arthritis (RA) involves several classes of pathogenic autoantibodies, some of which react with type-II collagen (COL2) in articular cartilage. We previously described a subset of COL2 antibodies targeting the F4 epitope (ERGLKGHRGFT) that could be regulatory. Here, using phage display, we developed recombinant antibodies against this epitope and examined the underlying mechanism of action. One of these antibodies, R69-4, protected against cartilage antibody- and collagen-induced arthritis in mice, but not autoimmune disease models independent of arthritogenic autoantibodies. R69-4 was further shown to cross-react with a large range of proteins within the inflamed synovial fluid, such as the complement protein C1q. Complexed R69-4 inhibited neutrophil FCGR3 signaling, thereby impairing downstream IL-1β secretion and neutrophil self-orchestrated recruitment. Likewise, human isotypes of R69-4 protected against arthritis with comparable efficiency. We conclude that R69-4 abrogates autoantibody-mediated arthritis mainly by hindering FCGR3 signaling, highlighting its potential clinical utility in acute RA.
Collapse
Affiliation(s)
- Zhongwei Xu
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Bingze Xu
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Susanna L Lundström
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Àlex Moreno-Giró
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
- Redoxis AB, Lund, Sweden
| | - Danxia Zhao
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Myriam Martin
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Erik Lönnblom
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Qixing Li
- Center for Medical Immunopharmacology Research, Southern Medical University, Guangzhou, China
| | - Alexander Krämer
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Changrong Ge
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Lei Cheng
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Bibo Liang
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
- Center for Medical Immunopharmacology Research, Southern Medical University, Guangzhou, China
| | - Dongmei Tong
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Roma Stawikowska
- Institute for Human Health & Disease Intervention and Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL, USA
| | - Anna M Blom
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Gregg B Fields
- Institute for Human Health & Disease Intervention and Department of Chemistry & Biochemistry, Florida Atlantic University, Jupiter, FL, USA
| | - Roman A Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Rikard Holmdahl
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
2
|
Schulz K, Trendelenburg M. C1q as a target molecule to treat human disease: What do mouse studies teach us? Front Immunol 2022; 13:958273. [PMID: 35990646 PMCID: PMC9385197 DOI: 10.3389/fimmu.2022.958273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
The complement system is a field of growing interest for pharmacological intervention. Complement protein C1q, the pattern recognition molecule at the start of the classical pathway of the complement cascade, is a versatile molecule with additional non-canonical actions affecting numerous cellular processes. Based on observations made in patients with hereditary C1q deficiency, C1q is protective against systemic autoimmunity and bacterial infections. Accordingly, C1q deficient mice reproduce this phenotype with susceptibility to autoimmunity and infections. At the same time, beneficial effects of C1q deficiency on disease entities such as neurodegenerative diseases have also been described in murine disease models. This systematic review provides an overview of all currently available literature on the C1q knockout mouse in disease models to identify potential target diseases for treatment strategies focusing on C1q, and discusses potential side-effects when depleting and/or inhibiting C1q.
Collapse
Affiliation(s)
- Kristina Schulz
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
- *Correspondence: Kristina Schulz,
| | - Marten Trendelenburg
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
3
|
Fitzpatrick EA, Wang J, Strome SE. Engineering of Fc Multimers as a Protein Therapy for Autoimmune Disease. Front Immunol 2020; 11:496. [PMID: 32269572 PMCID: PMC7109252 DOI: 10.3389/fimmu.2020.00496] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
The success of Intravenous Immunoglobulin in treating autoimmune and inflammatory processes such as immune thrombocytopenia purpura and Kawasaki disease has led to renewed interest in developing recombinant molecules capable of recapitulating these therapeutic effects. The anti-inflammatory properties of IVIG are, in part, due to the Fc region of the IgG molecule, which interacts with activating or inhibitory Fcγ receptors (FcγRs), the neonatal Fc Receptor, non-canonical FcRs expressed by immune cells and complement proteins. In most cases, Fc interactions with these cognate receptors are dependent upon avidity—avidity which naturally occurs when polyclonal antibodies recognize unique antigens on a given target. The functional consequences of these avid interactions include antibody dependent cell-mediated cytotoxicity, antibody dependent cell phagocytosis, degranulation, direct killing, and/or complement activation—all of which are associated with long-term immunomodulatory effects. Many of these immunologic effects can be recapitulated using recombinant or non-recombinant approaches to induce Fc multimerization, affording the potential to develop a new class of therapeutics. In this review, we discuss the history of tolerance induction by immune complexes that has led to the therapeutic development of artificial Fc bearing immune aggregates and recombinant Fc multimers. The contribution of structure, aggregation and N-glycosylation to human IgG: FcγR interactions and the functional effect(s) of these interactions are reviewed. Understanding the mechanisms by which Fc multimers induce tolerance and attempts to engineer Fc multimers to target specific FcγRs and/or specific effector functions in autoimmune disorders is explored in detail.
Collapse
Affiliation(s)
- Elizabeth A Fitzpatrick
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States
| | - Jin Wang
- College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| | - S E Strome
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States
| |
Collapse
|
4
|
Beaumont M, Tomazela D, Hodges D, Ermakov G, Hsieh E, Figueroa I, So OY, Song Y, Ma H, Antonenko S, Mengesha W, Zhang YW, Zhang S, Hseih S, Ayanoglu G, Du X, Rimmer E, Judo M, Vives F, Yearley JH, Moon C, Manibusan A, Knudsen N, Beck A, Bresson D, Gately D, Neupane D, Escandón E. Antibody-drug conjugates: integrated bioanalytical and biodisposition assessments in lead optimization and selection. AAPS OPEN 2018. [DOI: 10.1186/s41120-018-0026-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
5
|
Ho NI, Camps MGM, de Haas EFE, Trouw LA, Verbeek JS, Ossendorp F. C1q-Dependent Dendritic Cell Cross-Presentation of In Vivo-Formed Antigen-Antibody Complexes. THE JOURNAL OF IMMUNOLOGY 2017; 198:4235-4243. [PMID: 28432146 DOI: 10.4049/jimmunol.1602169] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 03/29/2017] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are specialized in Ag engulfment via a wide variety of uptake receptors on their cell surface. In the present study we investigated Ag uptake and presentation of in vivo-formed Ag-Ab complexes by i.v. injecting mice with Ag-specific Abs followed by the cognate Ag. We show by this natural Ab-mediated Ag targeting system that uptake by splenic APC subsets is severely hampered in mice lacking complement factor C1q (C1qa-/-). Moreover, no detectable Ag cross-presentation by CD8α+ DCs from C1qa-/- mice was found. On the contrary, Ag uptake was not hampered by APCs in FcγRI/II/III/IV-deficient (FcγR quadruple-/-) mice, and the cross-presentation ability of CD8α+ DCs was not affected. In conclusion, we show that C1q rather than FcγRs controls the Ab-mediated Ag uptake and its presentation by spleen APC subsets to T cells.
Collapse
Affiliation(s)
- Nataschja I Ho
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Marcel G M Camps
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Edwin F E de Haas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Leendert A Trouw
- Department of Rheumatology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands; and
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, the Netherlands;
| |
Collapse
|
6
|
Shen L, Engelhardt JA, Hung G, Yee J, Kikkawa R, Matson J, Tayefeh B, Machemer T, Giclas PC, Henry SP. Effects of Repeated Complement Activation Associated with Chronic Treatment of Cynomolgus Monkeys with 2'-O-Methoxyethyl Modified Antisense Oligonucleotide. Nucleic Acid Ther 2016; 26:236-49. [PMID: 27140858 DOI: 10.1089/nat.2015.0584] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The effects of repeated complement activation in cynomolgus monkeys after chronic antisense oligonucleotide (ASO) treatment were evaluated by using ISIS 104838, a representative 2'-O-methoxyethyl (2'-MOE) modified ASO. The treatment was up to 9 months with a total weekly dose of 30 mg/kg, given either as daily [4.3 mg/kg/day, subcutaneous (s.c.) injection] or once weekly [30 mg/kg, either as s.c. injection or 30-min intravenous (i.v.) infusion]. Acute elevations of complement split products (Bb and C3a) and a transient decrease in C3 occurred after the first dose and were drug plasma concentration dependent. However, with repeated complement activation after chronic ASO treatment, there were progressive increases in basal (predose) levels of Bb and C3a, and a sustained C3 reduction in all treated groups. There was also a progressive increase in C3d-bound circulating immune complex (CIC) that was considered secondary to the C3 depletion. Evidence of vascular inflammation was observed, mostly in the liver, kidney, and heart, and correlated with severe C3 depletion and increases in plasma IgG and IgM. Vascular inflammation was accompanied by increased C3 and IgM immunereactivity in the affected vasculatures and endothelial activation markers in serum. In summary, repeated complement activations in monkeys lead to a sustained decrease in circulating C3 over time. The concomitantly increased inflammatory signals and decreased CIC clearance due to impairment of complement function may lead to vascular inflammation after chronic ASO treatment in monkeys. However, based on the known sensitivity of monkeys to ASO-induced complement activation, these findings have limited relevance to humans.
Collapse
Affiliation(s)
- Lijiang Shen
- 1 Ionis Pharmaceuticals, Inc. , Carlsbad, California
| | | | - Gene Hung
- 1 Ionis Pharmaceuticals, Inc. , Carlsbad, California
| | - Jenna Yee
- 2 SNBL, USA, Ltd. , Everett, Washington
| | | | - John Matson
- 1 Ionis Pharmaceuticals, Inc. , Carlsbad, California
| | - Bryan Tayefeh
- 1 Ionis Pharmaceuticals, Inc. , Carlsbad, California
| | - Todd Machemer
- 1 Ionis Pharmaceuticals, Inc. , Carlsbad, California
| | | | - Scott P Henry
- 1 Ionis Pharmaceuticals, Inc. , Carlsbad, California
| |
Collapse
|
7
|
Beurskens FJ, van Schaarenburg RA, Trouw LA. C1q, antibodies and anti-C1q autoantibodies. Mol Immunol 2015; 68:6-13. [PMID: 26032012 DOI: 10.1016/j.molimm.2015.05.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/04/2015] [Accepted: 05/07/2015] [Indexed: 12/21/2022]
Abstract
The complement system has long been known for its role in combating infections. More recently the complement system is becoming increasingly appreciated for its role in processes that range from waste transport, immune tolerance and shaping of the adaptive immune response. Antibodies represent the humoral part of the adaptive immune response and the complement system interacts with antibodies in several ways. Activated complement fragments impact on the production of antibodies, the complement system gets activated by antibodies and complement proteins can be the target of (auto)antibodies. In this review, written to celebrate the contributions of Prof. Dr. M.R. Daha to the field of immunology and especially complement, we will focus on C1q and its various interactions with antibodies. We will specifically focus on the mechanisms by which C1q will interact with monomeric IgG versus polymerized IgG and fluid-phase IgM versus solid-phase IgM. In addition in this review we will discuss in detail how C1q itself is targeted by autoantibodies and how these autoantibodies are currently considered to play a role in human disease.
Collapse
Affiliation(s)
| | | | - Leendert A Trouw
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
8
|
Kottyan L, Kelly JA, Harley JB. Genetics of lupus. Rheumatology (Oxford) 2015. [DOI: 10.1016/b978-0-323-09138-1.00127-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
9
|
Fernandez-Arias C, Lopez JP, Hernandez-Perez JN, Bautista-Ojeda MD, Branch O, Rodriguez A. Malaria inhibits surface expression of complement receptor 1 in monocytes/macrophages, causing decreased immune complex internalization. THE JOURNAL OF IMMUNOLOGY 2013; 190:3363-72. [PMID: 23440418 DOI: 10.4049/jimmunol.1103812] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Complement receptor 1 (CR1) expressed on the surface of phagocytic cells binds complement-bound immune complexes (IC), playing an important role in the clearance of circulating IC. This receptor is critical to prevent accumulation of IC, which can contribute to inflammatory pathology. Accumulation of circulating IC is frequently observed during malaria, although the factors contributing to this accumulation are not clearly understood. We have observed that the surface expression of CR1 on monocytes/macrophages and B cells is strongly reduced in mice infected with Plasmodium yoelii, a rodent malaria model. Monocytes/macrophages from these infected mice present a specific inhibition of complement-mediated internalization of IC caused by the decreased CR1 expression. Accordingly, mice show accumulation of circulating IC and deposition of IC in the kidneys that inversely correlate with the decrease in CR1 surface expression. Our results indicate that malaria induces a significant decrease on surface CR1 expression in the monocyte/macrophage population that results in deficient internalization of IC by monocytes/macrophages. To determine whether this phenomenon is found in human malaria patients, we have analyzed 92 patients infected with either P. falciparum (22 patients) or P. vivax (70 patients) , the most prevalent human malaria parasites. The levels of surface CR1 on peripheral monocytes/macrophages and B cells of these patients show a significant decrease compared with uninfected control individuals in the same area. We propose that this decrease in CR1 plays an essential role in impaired IC clearance during malaria.
Collapse
Affiliation(s)
- Cristina Fernandez-Arias
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine, New York, NY 10010, USA
| | | | | | | | | | | |
Collapse
|
10
|
Abstract
Mouse models of lupus have for many years provided accessible and reliable research systems for the pathogenesis and therapy of systemic autoimmune disease, spanning a spectrum of inbred strains that develop spontaneous disease to experimentally induced, sometimes genetically manipulated animals. Nearly all the models share in common the development of glomerulonephritis and autoantibodies, including antinuclear and DNA specificities, the most common endpoints examined in experimental studies, but exhibit specific differences in the incidence of other end-organ manifestations such as hemolytic anemia, arthritis, dermatitis, and vasculitis. This chapter contrasts the clinical characteristics of these various models, providing an outline for their use and analysis.
Collapse
Affiliation(s)
- Stanford L Peng
- Rheumatology Clinical Research Unit, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| |
Collapse
|
11
|
Stoyanova V, Petrova S, Tchorbadjieva M, Deliyska B, Vasilev V, Tsacheva I. New insight into the autoimmunogenicity of the complement protein C1q. Mol Immunol 2010; 48:678-82. [PMID: 21159384 DOI: 10.1016/j.molimm.2010.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 10/06/2010] [Accepted: 11/16/2010] [Indexed: 10/18/2022]
Abstract
C1q along with its physiological role in maintenance of homeostasis and normal function of the immune system is involved in pathological conditions associated with repetitive generation of anti-C1q autoantibodies. The time and events that cause their first appearance are still unknown. We addressed this issue by analyzing the immunogenicity of C1q in two target groups-one of non-diseased humans and the other of lupus nephritis (LN) patients whose autoimmune disorder is associated with high titers of anti-C1q autoantibodies. The non-diseased humans were represented by pregnant women because the sex hormones are thought to be involved in triggering autoimmune pathologies by their ability to tip the balance of female adaptive immune response to production of antibodies. We screened, using ELISA, 31 sera from healthy pregnant women for the presence of IgM and IgG classes of autoantibodies, recognizing epitopes within the native C1q molecule, its collagen-like region (CLR) and globular head fragment (gC1q). The latter was represented by recombinant analogs of the three globular fragments of A, B and C chains, comprising C1q-ghA, ghB and ghC. We did not find IgM antibodies for all test-antigens which suggest that the natural IgM antibodies are not involved in triggering autoimmunity to C1q. Still more, we did not detect anti-CLR antibodies which have been proved pathogenic in already manifested LN. We completed the analysis with comparative epitope mapping of gC1q and we found similar immunogenic behavior in both target groups-ghA and ghC contained the immunodominant epitopes. This implies that the initial immune response to C1q might occur when the molecule has interacted with its ligands via ghB as part of gC1q. The presence of anti-gC1q in both healthy and diseased humans also implies that these antibodies, unlike anti-CLR, may have a contribution to an onset of autoimmunity.
Collapse
Affiliation(s)
- Vishnya Stoyanova
- Department of Biochemistry, Faculty of Biology, Sofia University, 8 Dragan Tsankov Str, 1164 Sofia, Bulgaria
| | | | | | | | | | | |
Collapse
|
12
|
Santer DM, Hall BE, George TC, Tangsombatvisit S, Liu CL, Arkwright PD, Elkon KB. C1q deficiency leads to the defective suppression of IFN-alpha in response to nucleoprotein containing immune complexes. THE JOURNAL OF IMMUNOLOGY 2010; 185:4738-49. [PMID: 20844193 DOI: 10.4049/jimmunol.1001731] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Almost all humans with homozygous deficiency of C1q develop systemic lupus erythematosus (SLE). The precise cellular mechanism(s) by which C1q prevents the development of SLE remains unclear. In this study, we tested the role of C1q in the regulation of IFN-α induced by immune complexes (ICs) in vitro, as well as the consequences of lack of C1q in vivo. Our experiments revealed that C1q preferentially promotes the binding of SLE ICs to monocytes rather than plasmacytoid dendritic cells, but this inhibition was not due to the induction of inhibitory soluble factors. The presence of C1q also altered the trafficking of ICs within monocytes such that ICs persisted in early endosomes. In patients with C1q deficiency, serum and cerebrospinal fluid levels of IFN-α and IFN-γ-inducible protein-10 levels were elevated and strongly correlated with Ro autoantibodies, demonstrating the clinical significance of these observations. These studies therefore associate C1q deficiency with defective regulation of IFN-α and provide a better understanding of the cellular mechanisms by which C1q prevents the development of IC-stimulated autoimmunity.
Collapse
Affiliation(s)
- Deanna M Santer
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Arazi A, Neumann AU. Modeling immune complex-mediated autoimmune inflammation. J Theor Biol 2010; 267:426-36. [PMID: 20832412 DOI: 10.1016/j.jtbi.2010.08.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 08/25/2010] [Accepted: 08/27/2010] [Indexed: 12/27/2022]
Abstract
A number of autoimmune diseases are thought to feature a particular type of self-sustaining inflammation, caused by the deposition of immune complexes (IC) in the inflamed tissue and a consequent activation of local effector cells. The persistence of this inflammation is due to a positive feedback loop, where autoantigen particles released as part of the tissue damage caused by the inflammation stimulate autoreactive B cells, leading to the formation of further immune complexes and their subsequent deposition. We present a mathematical model for the exploration of IC-mediated autoimmune inflammation and its clinical implications. We characterize the possible differences between normal individuals and those susceptible to such inflammation, and show that both random perturbations and bifurcations can lead to disease onset. Our model explains how defects in the mechanisms responsible for cellular debris clearance contribute to the development of disease, in agreement with empirical evidence. Moreover, we show that parameters governing the dynamics of immune complexes, such as their clearance rate, have an even stronger effect in determining the behavior of the system. We demonstrate the existence of hysteresis, implying that once IC-mediated autoimmune inflammation is triggered, its long-term suppression may be difficult to achieve. Our results can serve to guide the development of novel therapies to autoimmune diseases involving this type of inflammation.
Collapse
Affiliation(s)
- A Arazi
- Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel.
| | | |
Collapse
|
14
|
Tsirogianni A, Pipi E, Soufleros K. Relevance of anti-C1q autoantibodies to lupus nephritis. Ann N Y Acad Sci 2009; 1173:243-51. [PMID: 19758158 DOI: 10.1111/j.1749-6632.2009.04750.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The first component of the classical pathway of the complement system (C1q) is considered to have a crucial role in the clearance of immune complexes (ICs) as well as in the removal of waste material originating from apoptotic cells. A prolonged exposure of C1q epitopes to the immune system could eventually lead to an autoimmune response against itself. Although autoantibodies against C1q are found in several diseases, their clinical interest originates from their strong association to active lupus nephritis (LN). Several studies indicate that anti-C1q autoantibodies could serve as a reliable serologic marker in the assessment of LN activity compared to other immunological tests. Additionally, it was suggested that anti-C1q autoantibodies could play a role in LN pathogenesis. Their potential pathogenic actions likely depend on genetic background, titers, Ig classes and subclasses, and specific epitopes of anti-C1q autoantibodies as well as C1q availability and allocation. It is still unclear which different types of anti-C1q autoantibodies dominate in each case and if their upregulation is pathogenic, an epiphenomenon of aberrant tissue damage, or compensatory to an uncontrolled immune response.
Collapse
Affiliation(s)
- Alexandra Tsirogianni
- Department of Immunology-Histocompatibility, Evangelismos General Hospital, Athens, Greece.
| | | | | |
Collapse
|
15
|
Erdei A, Isaák A, Török K, Sándor N, Kremlitzka M, Prechl J, Bajtay Z. Expression and role of CR1 and CR2 on B and T lymphocytes under physiological and autoimmune conditions. Mol Immunol 2009; 46:2767-73. [PMID: 19559484 DOI: 10.1016/j.molimm.2009.05.181] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 05/29/2009] [Indexed: 11/17/2022]
Abstract
The involvement of complement in the development and regulation of antibody responses under both healthy and pathological conditions is known for long. Unravelling the molecular mechanisms underlying the events however is still in progress. This review focuses on the role of complement receptors CR1 (CD35) and CR2 (CD21) expressed on T and B cells. Alteration in the expression and function of these receptors may contribute to the initiation and maintenance of immune complex mediated autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis. Recent data regarding complement receptor expression on T lymphocytes and on memory B cells are also discussed.
Collapse
Affiliation(s)
- Anna Erdei
- Department of Immunology, Biological Institute, Eötvös Loránd University, Budapest, Hungary.
| | | | | | | | | | | | | |
Collapse
|
16
|
Avirutnan P, Mehlhop E, Diamond MS. Complement and its role in protection and pathogenesis of flavivirus infections. Vaccine 2009; 26 Suppl 8:I100-7. [PMID: 19388173 PMCID: PMC2768071 DOI: 10.1016/j.vaccine.2008.11.061] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The complement system is a family of serum and cell surface proteins that recognize pathogen-associated molecular patterns, altered-self ligands, and immune complexes. Activation of the complement cascade triggers several antiviral functions including pathogen opsonization and/or lysis, and priming of adaptive immune responses. In this review, we will examine the role of complement activation in protection and/or pathogenesis against infection by Flaviviruses, with an emphasis on experiments with West Nile and Dengue viruses.
Collapse
Affiliation(s)
- Panisadee Avirutnan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States
| | | | | |
Collapse
|
17
|
Gene expression profiles of European wild boar naturally infected with Mycobacterium bovis. Vet Immunol Immunopathol 2008; 129:119-25. [PMID: 19131115 DOI: 10.1016/j.vetimm.2008.12.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2008] [Revised: 12/01/2008] [Accepted: 12/08/2008] [Indexed: 11/20/2022]
Abstract
Global gene expression profiles were analyzed in European wild boar naturally infected with Mycobacterium bovis. Spleen RNA was extracted from 23 M. bovis-infected and 17 uninfected animals and analyzed using a Pigoligoarray representing 20,400 genes. Differentially expressed sequences (N=161) were identified affecting cellular processes such as apoptosis, cell communication and signal transduction, cell growth and/or maintenance, cytoskeleton organization and biogenesis, DNA repair, immune response, metabolism and energy pathways, protein metabolism, regulation of cell proliferation, regulation of gene expression, regulation of nucleic acid metabolism, regulation of physiological processes, and transport. Real-time RT-PCR analysis of mRNA levels was used to corroborate microarray results of selected genes. Immune response genes were among the most represented differentially expressed sequences and were selected for further discussion. Beta-defensin 129, T-cell surface glycoprotein CD8 and B-cell receptor-associated protein 29 were overexpressed in infected animals. Lower expression levels of the immune response genes galectin-1, complement component C1qB and certain HLA class I and class II histocompatibility antigens and immunoglobulin chains were found in infected animals. This study identified new mechanisms by which naturally infected European wild boar respond to M. bovis infection and how the pathogen circumvents host immune responses to establish infection. Gene expression studies in naturally infected wildlife reservoirs of bovine tuberculosis are important for functional genomics and vaccine studies to aid in disease control in wildlife.
Collapse
|
18
|
B cells in glomerulonephritis: focus on lupus nephritis. Semin Immunopathol 2007; 29:337-53. [PMID: 17943287 DOI: 10.1007/s00281-007-0092-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Accepted: 09/28/2007] [Indexed: 01/10/2023]
Abstract
The production of pathogenic antibody has been traditionally viewed as the principle contribution of B cells to the pathogenesis of immune-mediated glomerulonephritis. However, it is increasingly appreciated that B cells play a much broader role in such diseases, functioning as antigen-presenting cells, regulators of T cells, dendritic cells, and macrophages and orchestrators of local lymphatic expansion. In this review, we provide an overview of basic B cell biology and consider the evidence implicating B cells in one of the archetypal immune-mediated glomerulonephritides, lupus nephritis.
Collapse
|
19
|
Finke D, Randers K, Hoerster R, Hennig H, Zawatzky R, Marion T, Brockmann C, Klempt-Giessing K, Jacobsen K, Kirchner H, Goerg S. Elevated levels of endogenous apoptotic DNA and IFN-alpha in complement C4-deficient mice: implications for induction of systemic lupus erythematosus. Eur J Immunol 2007; 37:1702-9. [PMID: 17506029 DOI: 10.1002/eji.200636719] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Systemic lupus erythematosus (SLE), an autoimmune disease characterized by chronic nephritis, arthritis and dermatitis, and the presence of antinuclear autoantibodies, is associated with complement factor deficiencies in the classical activation pathway. In addition, IFN-alpha seems to be a key cytokine in SLE as an activated IFN-alpha system is regularly observed in patients with SLE. Here, we demonstrate that in lupus-susceptible, complement C4-deficient mice the lack of complement results in elevated intravascular levels of apoptotic DNA. The apoptotic DNA is targeted to the splenic marginal zone where it accumulates and induces IFN-alpha. As such, we present here a unifying hypothesis for the induction of SLE that incorporates the role of complement deficiency and elevated levels of IFN-alpha.
Collapse
MESH Headings
- Animals
- Antibodies, Antinuclear/immunology
- Antibodies, Antinuclear/metabolism
- Antibodies, Antinuclear/pharmacology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Antigen-Antibody Complex/analysis
- Antigen-Antibody Complex/immunology
- Apoptosis/genetics
- Apoptosis/immunology
- CD11b Antigen/analysis
- Complement C4/deficiency
- Complement C4/genetics
- DNA/immunology
- DNA/metabolism
- DNA/pharmacology
- Flow Cytometry
- Gene Expression/drug effects
- Immunoglobulin M/immunology
- Immunoglobulin M/metabolism
- Immunoglobulin M/pharmacology
- In Situ Nick-End Labeling
- Interferon-alpha/genetics
- Interferon-alpha/metabolism
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Leukocyte Common Antigens/analysis
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
Collapse
Affiliation(s)
- Doreen Finke
- Institute of Immunology and Transfusion Medicine, University of Lübeck, Lübeck, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
van Montfoort N, de Jong JMH, Schuurhuis DH, van der Voort EIH, Camps MGM, Huizinga TWJ, van Kooten C, Daha MR, Verbeek JS, Ossendorp F, Toes REM. A novel role of complement factor C1q in augmenting the presentation of antigen captured in immune complexes to CD8+ T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2007; 178:7581-6. [PMID: 17548593 DOI: 10.4049/jimmunol.178.12.7581] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ag-IgG immune complexes (IC) are efficiently taken up, and Ag-derived peptides are subsequently processed and presented by APC. In vitro experiments indicate that IgG Fc Receptors (FcgammaR) facilitate the efficient uptake of IC by dendritic cells. Previous experiments showed that the cross-presentation of Ag-derived peptides after s.c. administration of IC is FcgammaR-dependent. To study the role of different FcgammaR and complement in MHC class I Ag presentation after i.v. administration, we used mice deficient for FcgammaRs and complement components. These mice were injected with CFSE-labeled OVA-specific CD8+ T cells followed by administration of IC composed of OVA and rabbit anti-OVA IgG i.v. to measure MHC class I presentation of OVA-derived peptides. The Ag presentation was partly reduced in FcRgamma-chain-deficient mice, but not affected in FcgammaRI/II/III-deficient mice, complement factor C3-deficient mice, or FcgammaRI/II/III x C3-deficient mice. Importantly, CD8+ T cell proliferation was significantly reduced in mice deficient for C1q. This proliferation could be restored when IC were incubated with purified human C1q before injection. Likewise, purified C1q could strongly enhance the uptake and presentation of IC by dendritic cells in vitro. Heat inactivation abrogated the C1q-mediated uptake of IC. In addition, in vivo uptake of OVA-IC in the spleen was significantly reduced in C1q-deficient mice compared with wild-type mice. Together, these results indicate a novel function of C1q, which is present in high levels in the bloodstream, by directly enhancing the uptake and MHC class I presentation of Ag captured in IC by APC to CD8+ T cells.
Collapse
Affiliation(s)
- Nadine van Montfoort
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hepburn AL, Mason JC, Wang S, Shepherd CJ, Florey O, Haskard DO, Davies KA. Both Fcgamma and complement receptors mediate transfer of immune complexes from erythrocytes to human macrophages under physiological flow conditions in vitro. Clin Exp Immunol 2006; 146:133-45. [PMID: 16968408 PMCID: PMC1809732 DOI: 10.1111/j.1365-2249.2006.03174.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Abnormal clearance by the mononuclear phagocytic system of immune complexes (IC) is important in the pathogenesis of systemic lupus erythematosus (SLE). We have developed an in vitro model to investigate the cellular mechanisms involved in the transfer of soluble IC from erythrocytes to human macrophages under physiological flow conditions. In this assay, erythrocytes bearing fluorescently labelled IC are perfused over monolayers of human monocytes or monocyte-derived macrophages in a parallel-plate flow chamber, and transfer quantified using confocal microscopy and flow cytometry. Using aggregated human IgG as a model IC, we have been able to demonstrate transfer of IC from erythrocytes to macrophages. Blocking studies with specific neutralizing antibodies have shown that both complement and Fcgamma receptors are required for IC transfer. Blockade of CR4 (alpha(x)beta(2) integrin), FcgammaRIIa or FcgammaRIII reduced transfer, while anti-CR3 (alpha(m)beta(2) integrin) had no effect. Blockade of CR3, FcgammaRIIa or FcgammaRIII also reduced the number of adhesive interactions between fluorescently labelled IC-bearing erythrocytes and macrophage monolayers. Taken together with the transfer data, this suggests differing roles for these receptors in the human IC transfer reaction that includes an adhesive function which facilitates IC processing by mononuclear phagocytes. Finally, a functional effect of the FcgammaRIIa R131/H131 polymorphism, important in susceptibility to SLE, has also been demonstrated using this model. Uptake of IgG(2) but not IgG(1)-containing soluble IC was reduced by macrophages from individuals homozygous for the R131 allelic variant of the receptor.
Collapse
Affiliation(s)
- A L Hepburn
- Rheumatology Section, The Eric Bywaters Centre, Imperial College London, Hammersmith Hospital, London, UK.
| | | | | | | | | | | | | |
Collapse
|
22
|
de Jong JMH, Schuurhuis DH, Ioan-Facsinay A, Welling MM, Camps MGM, van der Voort EIH, Huizinga TWJ, Ossendorp F, Verbeek JS, Toes REM. Dendritic cells, but not macrophages or B cells, activate major histocompatibility complex class II-restricted CD4+ T cells upon immune-complex uptake in vivo. Immunology 2006; 119:499-506. [PMID: 16995881 PMCID: PMC2265814 DOI: 10.1111/j.1365-2567.2006.02464.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Professional antigen-presenting cells (APC) are able to process and present exogenous antigen leading to the activation of T cells. Antigen-immunoglobulin (Ig)G complexes (IC) are much more efficiently processed and presented than soluble antigen. Dendritic cells (DC) are known for their ability to take up and process immune complex (IC) via FcgammaR, and they have been shown to play a crucial role in IC-processing onto major histocompatibility complex (MHC) class I as they contain a specialized cross-presenting transport system required for MHC class I antigen-processing. However, the MHC class II-antigen-processing pathway is distinct. Therefore various other professional APC, like macrophages and B cells, all displaying FcgammaR, are thought to present IC-delivered antigen in MHC class II. Nonetheless, the relative contribution of these APC in IC-facilitated antigen-presentation for MHC class II in vivo is not known. Here we show that, in mice, both macrophages and DC, but not B cells, efficiently capture IC. However, only DC, but not macrophages, efficiently activate antigen-specific MHC class II restricted CD4(+) T cells. These results indicate that mainly DC and not other professional APC, despite expressing FcgammaR and MHC class II, contribute significantly to IC-facilitated T cell activation in vivo under steady-state conditions.
Collapse
Affiliation(s)
- Judith M H de Jong
- Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Sontheimer RD, Racila E, Racila DM. C1q: Its Functions within the Innate and Adaptive Immune Responses and its Role in Lupus Autoimmunity. J Invest Dermatol 2005; 125:14-23. [PMID: 15982298 DOI: 10.1111/j.0022-202x.2005.23673.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The complement cascade is a multi-faced effector component of the innate immune response. C1q is the recognition component of the classical pathway of complement activation. In addition, C1q has been recognized to serve a number of other biological functions including a modulating role on cellular functions within the adaptive immune response. The importance of C1q to normal immune regulation is reflected by the fact that greater than 90% of individuals who have complete congenital deficiency of C1q have been observed to develop early-onset photosensitive systemic lupus erythematosus (SLE). As a number of single nucleotide polymorphisms have been identified in three C1q genes, it is possible that more subtle variations in C1q expression could be a risk factor for cutaneous LE and SLE. Thus, a more comprehensive delineation of complotype could be of increasing clinical importance in the future.
Collapse
Affiliation(s)
- Richard D Sontheimer
- Department of Dermatology, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA.
| | | | | |
Collapse
|
24
|
Trendelenburg M, Manderson AP, Fossati-Jimack L, Walport MJ, Botto M. Monocytosis and accelerated activation of lymphocytes in C1q-deficient autoimmune-prone mice. Immunology 2004; 113:80-8. [PMID: 15312138 PMCID: PMC1782548 DOI: 10.1111/j.1365-2567.2004.01940.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
C1q deficiency has been shown to accelerate spontaneous autoimmunity in mice. We studied the time course of activation of monocytes and lymphocytes in autoimmune and non-autoimmune mice in the presence or absence of C1q as a disease accelerator. Autoimmune MRL\Mp.C1qa-\- and non-autoimmune C57BL\6.C1qa-\- mice were analysed at various time points between 6 and 33 weeks of age and compared to strain- and age-matched C1q-sufficient controls. Splenic and peritoneal leucocytes were analysed by flow cytometry and plasma levels of immunoglobulin M (IgM), total IgG, IgG subclasses and IgM autoantibodies were measured. Both C1q-deficient strains had significantly more splenic monocytes than their controls at all time points analysed. In addition, MRL\Mp.C1qa-\- but not C57BL/6.C1qa-\- mice developed splenic hypercellularity starting at about 12-17 weeks old, had signs of accelerated CD4+ T-cell activation and showed a marked increase in splenic plasma cells and total serum IgM levels from about 22 weeks of age. The accelerated CD4+ T-cell activation was not due to a direct inhibitory effect of C1q on T cells. These data show that C1q deficiency causes splenic monocytosis together with accelerated T-cell activation in an autoimmune-prone mouse strain.
Collapse
Affiliation(s)
- Marten Trendelenburg
- Rheumatology Section, Eric Bywaters Centre, Faculty of Medicine, Imperial College, Hammersmith Campus, London, UK
| | | | | | | | | |
Collapse
|
25
|
Manderson AP, Botto M, Walport MJ. The role of complement in the development of systemic lupus erythematosus. Annu Rev Immunol 2004; 22:431-56. [PMID: 15032584 DOI: 10.1146/annurev.immunol.22.012703.104549] [Citation(s) in RCA: 371] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Complement has both beneficial and deleterious roles in the pathogenesis of systemic lupus erythematosus (SLE). On the one hand, patients with SLE present with decreased complement levels and with complement deposition in inflamed tissues, suggestive of a harmful role of complement in the effector phase of disease. On the other hand, homozygous deficiency of any of the classical pathway proteins is strongly associated with the development of SLE. There are two main hypotheses to explain these observations. The first invokes an important role for complement in the physiological waste-disposal mechanisms of dying cells and immune complexes. The second hypothesis is based around the role of complement in determining the activation thresholds of B and T lymphocytes, with the proposal that complement deficiency causes incomplete maintenance of peripheral tolerance. These two hypotheses are not mutually exclusive. In addition, there is evidence for a contribution from other genetic factors in determining the phenotype of disease in the absence of complement.
Collapse
Affiliation(s)
- Anthony P Manderson
- Rheumatology Section, Division of Medicine, Faculty of Medicine, Imperial College, Hammersmith Campus, London W12 0NN, United Kingdom.
| | | | | |
Collapse
|
26
|
Wilber A, O'Connor TP, Lu ML, Karimi A, Schneider MC. Dnase1l3 deficiency in lupus-prone MRL and NZB/W F1 mice. Clin Exp Immunol 2003; 134:46-52. [PMID: 12974753 PMCID: PMC1808827 DOI: 10.1046/j.1365-2249.2003.02267.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Loss of deoxyribonuclease I (Dnase1) function is associated with systemic lupus erythematosus (SLE) in humans and mice; however, no coding mutations in Dnase1 are found in polygenic murine models. Instead, both MRL-lpr strains and NZB/W F1 hybrids are homozygous for T89I missense in the macrophage-DNASE, desoxyribonuclease I-like 3 (Dnase1l3). By in vitro expression studies, this substitution decreases this enzyme's nuclease activity against free DNA by only approximately twofold; however, the mutation has a greater effect on the capacity of media conditioned with Dnase1l3 to confer a barrier to liposomal gene transfection to HeLa cells. The 89I substitution decreases the Dnase1l3 barrier function in vitro by eightfold (P < 0.01). In splenocytes and BM-derived macrophages of SLE mice, while cellular Dnase1l3 levels are induced relative to C57BL/6 (control) mice, levels of FD-nuclease activity are similar. Finally, media conditioned by MRL and NZB/W F1 macrophages, relative to control, contains a weak interferon-gamma (IFN-gamma) inducible Dnase1l3-associated barrier to transfection. This barrier function is hypothesized to reflect the inability of SLE mice to degrade membrane-enveloped DNA-associated antigens, such as apoptotic bodies, which are predicted to stimulate the characteristic autoimmunity of SLE. Our results for these two generally independent models strongly suggest that Dnase1l3 deficiency increases the susceptibility of these mice to polygenic SLE.
Collapse
Affiliation(s)
- A Wilber
- Division of Genetics and Metabolism, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | | | | | | | | |
Collapse
|
27
|
Racila DM, Sontheimer CJ, Sheffield A, Wisnieski JJ, Racila E, Sontheimer RD. Homozygous single nucleotide polymorphism of the complement C1QA gene is associated with decreased levels of C1q in patients with subacute cutaneous lupus erythematosus. Lupus 2003; 12:124-32. [PMID: 12630757 DOI: 10.1191/0961203303lu329oa] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We report an association between a non-familial form of photosensitive Lupus-specific skin disease, subacute cutaneous lupus erythematosus (SCLE), and a new single nucleotide polymorphism (SNP) in the C1QA gene. We also describe an association between this SNP and lower levels of serum C1q. This SNP consists of adenine replacing the third guanine in the codon for aminoacid residue Gly70 (position excludes the 22 amino acid leading peptide) that is located in the second exon of the C1QA gene. We have designated this SNP C1qA-Gly70GGA (the GenBank sequence at this location is C1qA-Gly70GGG). A survey of 19 SCLE patients showed that 11 (58%) were homozygous for C1qA-Gly70GGA SNP, seven (37%) were heterozygous, and only one patient (5%) was homozygous for the GenBank sequence. In contrast, only 13 of 62 (21%) normal controls were homozygous for the C1qA-Gly70GGA SNP, 41 (66%) controls were heterozygous and eight (13%) controls were homozygous for the GenBank sequence. Thus, the C1qA-Gly70GGA SNP is strongly associated with SCLE (P-value = 0.005 by chi-square analysis with Yates correction). This SNP would traditionally be classified as clinically silent as it does not encode a different amino acid. However, our studies have suggested that this SNP appears to be associated with a functional abnormality of C1q expression since its presence correlates inversely with serum levels of C1q antigenic protein in both SCLE patients and normal controls. The mechanism by which this phenotypic change is associated with the translationally silent (synonymous) ClqA-Gly70GGA genetic variation is currently unknown.
Collapse
Affiliation(s)
- D M Racila
- Department of Dermatology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | | | | | | | | | | |
Collapse
|
28
|
O'Brien BA, Huang Y, Geng X, Dutz JP, Finegood DT. Phagocytosis of apoptotic cells by macrophages from NOD mice is reduced. Diabetes 2002; 51:2481-8. [PMID: 12145161 DOI: 10.2337/diabetes.51.8.2481] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Macrophages limit inflammatory responses by clearing apoptotic cells. Deficiencies in apoptotic cell phagocytosis have been linked to autoimmunity. In this study, we determined the efficiency with which macrophages from diabetes-prone NOD and diabetes-resistant NOR, Idd5, Balb/c, and C57BL/6 mice phagocytose apoptotic thymocytes and NIT-1 insulinoma cells. Peritoneal and bone marrow-derived macrophages from NOD mice engulfed fewer apoptotic thymocytes than macrophages from Balb/c mice (P < 0.05). Peritoneal macrophages from NOR and Idd5 NOD congenic mice were more proficient at engulfment than their NOD counterparts. Annexin V blockade diminished apoptotic thymocyte clearance and heat-labile serum factors augmented clearance. Binding of apoptotic thymocytes to NOD macrophages was also reduced, suggesting that the deficiency in phagocytosis may be partly attributable to a recognition defect. Peritoneal macrophages from female Balb/c and NOD mice were equally efficient in the engulfment of microspheres, suggesting that the phagocytic deficiency observed in NOD mice was specific for apoptotic cells. In summary, we have demonstrated a deficiency in phagocytic function of macrophages from NOD mice. Normal and diabetes-prone neonatal rodents have a wave of beta-cell apoptosis coincident with the onset of target organ inflammation. A constitutive defect in the clearance of apoptotic beta-cells may be contributory to the initiation of autoimmunity.
Collapse
Affiliation(s)
- Bronwyn A O'Brien
- Diabetes Research Laboratory, Department of Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | | | | | | |
Collapse
|
29
|
Abstract
Complement, an important component of the innate immune system, is comprised of about 35 individual proteins. In mammals, activation of complement results in the generation of activated protein fragments that play a role in microbial killing, phagocytosis, inflammatory reactions, immune complex clearance, and antibody production. Fish appear to possess activation pathways similar to those in mammals, and the fish complement proteins identified thus far show many homologies to their mammalian counterparts. Because information about complement proteins, regulatory proteins, and complement receptors in fish is far from complete, it is unclear whether all the complement functions that have been identified in mammals also occur in fish. However, it has been clearly demonstrated that fish complement can lyse foreign cells and opsonise foreign organisms for destruction by phagocytes. There are also indications that complement fragments participate in inflammatory reactions. Fish possess multiple isoforms of several complement proteins, such as C3 and factor B. It has been hypothesised that the function of this diversity in complement proteins serves to expand their innate immune recognition capacity and response. Understanding the functions of complement in fish and the roles the individual proteins, including the various isoforms, play in host defence, is important not only for understanding the evolution of this system but also for the development of new strategies in fish health management.
Collapse
Affiliation(s)
- M Claire H Holland
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 401 Stellar Chance Laboratories, Philadelphia, PA 19014, USA
| | | |
Collapse
|
30
|
Shlomchik MJ, Craft JE, Mamula MJ. From T to B and back again: positive feedback in systemic autoimmune disease. Nat Rev Immunol 2001; 1:147-53. [PMID: 11905822 DOI: 10.1038/35100573] [Citation(s) in RCA: 436] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Systemic lupus erythematosus, a prototypical systemic autoimmune disease, is the result of a series of interactions within the immune system that ultimately lead to the loss of self-tolerance to nuclear autoantigens. Here, we present an integrated model that explains how self-tolerance is initially lost and how the loss of tolerance is then amplified and maintained as a chronic autoimmune state. Key to this model are the self-reinforcing interactions of T and B cells, which we suggest lead to perpetuation of autoimmunity as well as its spread to multiple autoantigen targets.
Collapse
Affiliation(s)
- M J Shlomchik
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8035, USA
| | | | | |
Collapse
|
31
|
Manderson AP, Pickering MC, Botto M, Walport MJ, Parish CR. Continual low-level activation of the classical complement pathway. J Exp Med 2001; 194:747-56. [PMID: 11560991 PMCID: PMC2195964 DOI: 10.1084/jem.194.6.747] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
There is evidence that the classical complement pathway may be activated via a "C1-tickover" mechanism, analogous to the C3-tickover of the alternative pathway. We have quantitated and characterized this pathway of complement activation. Analysis of freshly collected mouse and human plasma revealed that spontaneous C3 activation rapidly occurred with the generation of C3 fragments in the plasma. By the use of complement- and Ig-deficient mice it was found that C1q, C4, C2, and plasma Ig were all required for this spontaneous C3 activation, with the alternative complement pathway further amplifying C3 fragment generation. Study of plasma from a human with C1q deficiency before and after therapeutic C1q infusion confirmed the existence of a similar pathway for complement activation in humans. Elevated levels of plasma C3 were detected in mice deficient in complement components required for activation of either the classical or alternative complement pathways, supporting the hypothesis that there is continuous complement activation and C3 consumption through both these pathways in vivo. Blood stasis was found to stimulate C3 activation by classical pathway tick-over. This antigen-independent mechanism for classical pathway activation may augment activation of the complement system at sites of inflammation and infarction.
Collapse
Affiliation(s)
- Anthony P. Manderson
- Division of Immunology and Cell Biology, John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - Matthew C. Pickering
- Rheumatology Section, Division of Medicine, Imperial College School of Medicine, London W12 ONN, UK
| | - Marina Botto
- Rheumatology Section, Division of Medicine, Imperial College School of Medicine, London W12 ONN, UK
| | - Mark J. Walport
- Rheumatology Section, Division of Medicine, Imperial College School of Medicine, London W12 ONN, UK
| | - Christopher R. Parish
- Division of Immunology and Cell Biology, John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| |
Collapse
|