Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Eur J Immunol ; 54(7): e2350832, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38700064

ABSTRACT

Seventy-seven patients with antiphospholipid syndrome were tested for autoantibodies against C1q, C3, FB, FH, and C4bp. Fifty-seven patients had at least one anti-complement antibody. IgM anti-FH positivity was associated with thrombosis when anti-C3 and anti-FB were, negatively or positively, associated with various noncriteria manifestations of antiphospholipid syndrome.


Subject(s)
Antiphospholipid Syndrome , Autoantibodies , Complement System Proteins , Humans , Antiphospholipid Syndrome/immunology , Autoantibodies/immunology , Autoantibodies/blood , Female , Male , Middle Aged , Adult , Complement System Proteins/immunology , Prevalence , Immunoglobulin M/immunology , Immunoglobulin M/blood , Thrombosis/immunology , Aged
2.
Am J Kidney Dis ; 82(3): 279-289, 2023 09.
Article in English | MEDLINE | ID: mdl-37061020

ABSTRACT

RATIONALE & OBJECTIVE: C3 glomerulopathy (C3GN) and atypical hemolytic uremic syndrome (aHUS) are 2 distinct rare kidney diseases caused by dysregulation of the alternative complement pathway. Patients with C3GN and concurrent kidney lesions of thrombotic microangiopathy (TMA) have been rarely reported. We characterized the clinical features and underlying immunological abnormalities in these patients. STUDY DESIGN: Case series. SETTING & PARTICIPANTS: Patients with C3GN and concomitant TMA lesions on biopsy registered from 2009 to 2019 in the French National Registry of C3GN. FINDINGS: Among 278 registered patients with C3GN, 16 (6%) had biopsy-proven glomerular and/or vascular TMA lesions. Their median age at diagnosis was 39 years (range, 7-76), and 59% were female. Fourteen of the 16 patients (88%) had an estimated glomerular filtration rate of<30mL/min/1.73m2 and 3 of 16 (19%) required dialysis. Twelve of the 14 evaluated patients (86%) showed evidence of mechanical hemolysis. Fifty percent of the patients had low C3 levels. Six of the 14 evaluated patients had a rare variant in complement genes, and 4 of the 16 patients (25%) had monoclonal gammopathy. Among the 16 patients, 10 (63%) received eculizumab, 5 (31%) received immunosuppressive therapy, and 4 (25%) received clone-targeted chemotherapy. Median kidney survival was 49 months. LIMITATIONS: Small retrospective case series with a limited number of biopsies including electron microscopy. CONCLUSIONS: Concomitant C3GN and TMA is extremely rare and is associated with poor kidney outcomes. Genetic or acquired abnormalities of the alternative complement pathway are common as is the presence of monoclonal gammopathy, which may inform the selection of treatment approaches.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Paraproteinemias , Thrombotic Microangiopathies , Humans , Female , Child , Adolescent , Young Adult , Adult , Middle Aged , Aged , Male , Retrospective Studies , Kidney , Atypical Hemolytic Uremic Syndrome/drug therapy , Thrombotic Microangiopathies/therapy , Thrombotic Microangiopathies/complications , Paraproteinemias/complications
3.
Int J Mol Sci ; 24(4)2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36834827

ABSTRACT

The interaction of some human antibodies with heme results in posttranslational acquisition of binding to various self- and pathogen-derived antigens. The previous studies on this phenomenon were performed with oxidized heme (Fe3+). In the present study, we elucidated the effect of other pathologically relevant species of heme, i.e., species that were formed after contact of heme with oxidizing agents such as hydrogen peroxide, situations in which heme's iron could acquire higher oxidation states. Our data reveal that hyperoxidized species of heme have a superior capacity to heme (Fe3+) in triggering the autoreactivity of human IgG. Mechanistic studies demonstrated that oxidation status of iron was of critical importance for the heme's effect on antibodies. We also demonstrated that hyperoxidized heme species interacted at higher affinities with IgG and that this binding occurred through a different mechanism as compared to heme (Fe3+). Regardless of their profound functional impact on the antigen-binding properties of antibodies, hyperoxidized species of heme did not affect Fc-mediated functions of IgG, such as binding to the neonatal Fc receptor. The obtained data contribute to a better understanding of the pathophysiological mechanism of hemolytic diseases and of the origin of elevated antibody autoreactivity in patients with some hemolytic disorders.


Subject(s)
Heme , Immunoglobulin G , Infant, Newborn , Humans , Heme/metabolism , Oxidation-Reduction , Adaptive Immunity , Iron
4.
Am J Kidney Dis ; 80(3): 341-352, 2022 09.
Article in English | MEDLINE | ID: mdl-35217094

ABSTRACT

RATIONALE & OBJECTIVE: Hemolytic uremic syndrome (HUS), a thrombotic microangiopathy (TMA) with kidney involvement, is a rare condition in patients with monoclonal gammopathy. In the absence of known causes of TMA, the role of complement activation in endothelial injury in patients with monoclonal gammopathy remains unknown and was the focus of this investigation. STUDY DESIGN: Case series. SETTING & PARTICIPANTS: We studied the 24 patients in the French national registry of HUS between 2000 and 2020 who had monoclonal gammopathy without other causes of secondary TMA. We provide the clinical histories and complement studies of these patients. FINDINGS: Monoclonal gammopathy-associated TMA with kidney involvement is estimated to be 10 times less frequent than adult atypical HUS (aHUS) in the French national registry. It is characterized by severe clinical features, with 17 of 24 patients requiring dialysis at disease onset, and with median renal survival of only 20 months. TMA-mediated extrarenal manifestations, particularly cutaneous and neurological involvement, were common and associated with poor overall prognosis. Complement studies identified low C3, normal C4, and high soluble C5b-9 levels in 33%, 100%, and 77% of tested patients, respectively, indicating a contribution of the alternative and terminal complement pathways in the pathophysiology of the disease. Genetic abnormalities in complement genes known to be associated with aHUS were found in only 3 of 17 (17%) who were tested. LIMITATIONS: Retrospective study without comparison group; limited number of patients, limited available blood samples. CONCLUSIONS: Within the spectrum of TMA, TMA associated with monoclonal gammopathy represents a distinct subset. Our findings suggest that HUS associated with monoclonal immunoglobulin is a complement-mediated disease akin to aHUS.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Paraproteinemias , Thrombotic Microangiopathies , Adult , Antibodies, Monoclonal, Humanized , Atypical Hemolytic Uremic Syndrome/epidemiology , Atypical Hemolytic Uremic Syndrome/genetics , Complement Activation , Complement System Proteins , Humans , Paraproteinemias/complications , Paraproteinemias/epidemiology , Retrospective Studies , Thrombotic Microangiopathies/epidemiology , Thrombotic Microangiopathies/etiology
5.
Clin Chem Lab Med ; 60(9): 1373-1383, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35749077

ABSTRACT

OBJECTIVES: No reference data are available on repositories to measure precision of autoantibody assays. The scope of this study was to document inter- and intra-run variations of quantitative autoantibody assays based on a real-world large international data set. METHODS: Members of the European Autoimmunity Standardisation Initiative (EASI) group collected the data of intra- and inter-run variability obtained with assays quantifying 15 different autoantibodies in voluntary participating laboratories from their country. We analyzed the impact on the assay performances of the type of immunoassay, the number of measurements used to calculate the coefficient of variation (CVs), the nature and the autoantibody level of the internal quality control (IQC). RESULTS: Data were obtained from 64 laboratories from 15 European countries between February and October 2021. We analyzed 686 and 1,331 values of intra- and inter-run CVs, respectively. Both CVs were significantly dependent on: the method of immunoassay, the level of IQC with higher imprecision observed when the antibody levels were lower than 2-fold the threshold for positivity, and the nature of the IQC with commercial IQCs having lower CVs than patients-derived IQCs. Our analyses also show that the type of autoantibody has low impact on the assay' performances and that 15 measurements are sufficient to establish reliable intra- and inter-run variations. CONCLUSIONS: This study provides for the first time an international repository yielding values of intra- and inter-run variation for quantitative autoantibody assays. These data could be useful for ISO 15189 accreditation requirements and will allow clinical diagnostic laboratories to assure quality of patient results.


Subject(s)
Autoantibodies , Clinical Laboratory Services , Humans , Laboratories , Quality Control , Reference Standards
6.
Pediatr Nephrol ; 37(7): 1479-1493, 2022 07.
Article in English | MEDLINE | ID: mdl-35118546

ABSTRACT

BACKGROUND AND OBJECTIVES: Observing biomarkers that affect alternative pathway dysregulation components may be effective in obtaining a new and more rapid diagnostic portrayal of atypical hemolytic uremic syndrome. We have conducted a systematic review on the aHUS biomarkers: C3, C5a, C5b-9, factor B, complement factor B, H, and I, CH50, AH50, D-dimer, as well as anti-CFH antibodies. METHODS: An exhaustive literature search was conducted for aHUS patient population plasma/serum, collected/reported at the onset of diagnosis. A total of 60 studies were included with the data on 837 aHUS subjects, with at least one biomarker reported. RESULTS: The biomarkers C3 [mean (SD): 72.1 (35.0), median: 70.5 vs. reference range: 75-175 mg/dl, n = 752]; CH50 [28.3 (32.1), 24.3 vs. 30-75 U/ml, n = 63]; AH50 [27.6% (30.2%), 10% vs. ≥ 46%, n = 23]; and CFB [13.1 (6.6), 12.4, vs. 15.2-42.3 mg/dl, n = 19] were lower among aHUS subjects as compared with the reference range. The biomarkers including C4 [mean (SD): 20.4 (9.5), median: 20.5 vs. reference range: 14-40 mg/dl, n = 343]; C4d [7.2 (6.5), 4.8 vs. ≤ 9.8 µg/ml, n = 108]; CFH [40.2 (132.3), 24.5 vs. 23.6-43.1 mg/dl, n = 123 subjects]; and CFI [8.05 (5.01), 6.55 mg/dl vs. 4.4-18.1 mg/dl, n = 38] were all observed to be within the reference range among aHUS subjects. The biomarkers C5a [mean (SD): 54.9 (32.9), median: 48.8 vs. reference range: 10.6-26.3 mg/dl, n = 117]; C5b-9 [466.0 (401.4), 317 (186-569.7) vs. ≤ 250 ng/ml, n = 174]; Bb [2.6 (2.1), 1.9 vs. ≤ 1.6 µg/ml, n = 77] and D-dimer [246 (65.05), 246 vs. < 2.2 ng/ml, 2, n = 2 subjects] were higher among patients with aHUS compared with the reference range. CONCLUSION: If a comprehensive complement profile were built using our data, aHUS would be identified by low levels of C3, CH50, AH50, and CFB along with increased levels of C5a, C5b-9, Bb, anti-CFH autoantibodies, and D-dimer. A higher resolution version of the Graphical abstract is available as Supplementary information.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Atypical Hemolytic Uremic Syndrome/diagnosis , Autoantibodies , Biomarkers , Complement Factor B , Complement Factor H , Complement Membrane Attack Complex , Humans
8.
Pediatr Nephrol ; 34(8): 1465-1482, 2019 08.
Article in English | MEDLINE | ID: mdl-30989342

ABSTRACT

BACKGROUND: Hemolytic uremic syndrome (HUS) is a leading cause of acute kidney injury in children. Although international guidelines emphasize comprehensive evaluation and treatment with eculizumab, access to diagnostic and therapeutic facilities is limited in most developing countries. The burden of Shiga toxin-associated HUS in India is unclear; school-going children show high prevalence of anti-factor H (FH) antibodies. The aim of the consensus meeting was to formulate guidelines for the diagnosis and management of HUS in children, specific to the needs of the country. METHODS: Four workgroups performed literature review and graded research studies addressing (i) investigations, biopsy, genetics, and differential diagnosis; (ii) Shiga toxin, pneumococcal, and infection-associated HUS; (iii) atypical HUS; and (iv) complement blockade. Consensus statements developed by the workgroups were discussed during a consensus meeting in March 2017. RESULTS: An algorithm for classification and evaluation was developed. The management of Shiga toxin-associated HUS is supportive; prompt plasma exchanges (PEX) is the chief therapy in patients with atypical HUS. Experts recommend that patients with anti-FH-associated HUS be managed with a combination of PEX and immunosuppressive medications. Indications for eculizumab include incomplete remission with plasma therapy, life-threatening features, complications of PEX or vascular access, inherited defects in complement regulation, and recurrence of HUS in allografts. Priorities for capacity building in regional and national laboratories are highlighted. CONCLUSIONS: Limited diagnostic capabilities and lack of access to eculizumab prevent the implementation of international guidelines for HUS in most developing countries. We propose practice guidelines for India, which will perhaps be applicable to other developing countries.


Subject(s)
Consensus Development Conferences as Topic , Hemolytic-Uremic Syndrome/diagnosis , Nephrology/standards , Practice Guidelines as Topic , Shiga-Toxigenic Escherichia coli/immunology , Consensus , Developing Countries , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/microbiology , Humans , India , Nephrology/methods , Plasma Exchange , Shiga-Toxigenic Escherichia coli/isolation & purification
10.
Pediatr Res ; 84(1): 118-124, 2018 07.
Article in English | MEDLINE | ID: mdl-29795200

ABSTRACT

BACKGROUND: Prodromal symptoms are frequently reported in the atypical form of Hemolytic uremic syndrome (aHUS) suggesting implication of infectious triggers. Some pathogens may also play a role in the mechanisms of production of autoantibody directed against Factor H (FH), a complement regulator, leading to aHUS. METHODS: The presence of 15 gastrointestinal (GI) pathogens was investigated by using xTAG-based multiplex PCR techniques on stools collected at the acute phase in a cohort of Indian HUS children classified according to the presence or absence of anti-FH autoantibodies. RESULTS: Prevalence of pathogens in patients with anti-FH antibody (62.5%) was twice that in those without (31.5%). Different pathogens were detected, the most frequent being Clostridium difficile, Giardia intestinalis, Salmonella, Shigella, Rotavirus, Norovirus and Entamoeba histolytica. No stool was positive for Shigatoxin. CONCLUSION: This study reveals a higher prevalence of GI pathogens in anti-FH positive than in negative patients. No single pathogen was implicated exclusively in one form of HUS. These pathogens may play a role in the disease initiation by inducing complement activation or an autoimmune response.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Autoantibodies/immunology , Complement Activation , Atypical Hemolytic Uremic Syndrome/microbiology , Atypical Hemolytic Uremic Syndrome/parasitology , Atypical Hemolytic Uremic Syndrome/virology , Child , Child, Preschool , Clostridioides difficile , Cohort Studies , Complement Factor H/immunology , Entamoeba histolytica , Female , Giardia lamblia , Humans , India , Infant , Intestines/microbiology , Intestines/parasitology , Intestines/pathology , Intestines/virology , Male , Multiplex Polymerase Chain Reaction , Mutation , Norovirus , Rotavirus , Salmonella , Shigella
11.
Kidney Int ; 92(5): 1232-1241, 2017 11.
Article in English | MEDLINE | ID: mdl-28712854

ABSTRACT

C3 Glomerulopathies, which include Dense Deposit Disease and C3 Glomerulonephritis, are associated with genetic and acquired dysregulation of the C3 convertase alternative pathway of complement. The potential role of the activation of the C5 convertase has not been studied extensively. Here we analyzed IgG samples from patients with C3 Glomerulopathies to identify circulating autoantibodies that stabilize the C3 alternative pathway (C3 Nephritic Factors) as well as C5 convertases (C5 Nephritic Factors), thus preventing decay of these enzyme complexes. Rare variants in alternative pathway genes were found in 28 of 120 tested patients. C3 and C5 Nephritic Factors were found in 76 of 101 (75%) and 29 of 59 (49%) of the patients, respectively. Therefore, we compared the results of the assays for the C3 and C5 nephritic factors functional activity: 29% were positive for C3 Nephritic Factors alone, 39% were positive for both C3 and C5 Nephritic Factors, and 10% were positive for C5 Nephritic Factors alone. We found that the addition of properdin-enhanced stabilization of C3 convertase in the presence of IgG doubly positive for both Nephritic Factors, while it did not modify the stabilization mediated by IgG solely positive for C3 Nephritic Factors. Both C3 and C5 Nephritic Factors correlated with C3 consumption, while only C5 Nephritic Factors correlated with sC5b9 levels. C5 Nephritic Factors-positive patients were more likely to have C3 Glomerulonephritis than Dense Deposit Disease. Thus, dysregulation of the C5 convertase contributes to C3 Glomerulopathies inter-disease differences and may have direct therapeutic implications.


Subject(s)
Complement C3 Convertase, Alternative Pathway/immunology , Complement C3 Nephritic Factor/immunology , Complement C3-C5 Convertases/immunology , Complement Pathway, Alternative/immunology , Glomerulonephritis, Membranoproliferative/immunology , Adolescent , Adult , Child , Complement C3 Nephritic Factor/analysis , Complement C3 Nephritic Factor/genetics , Complement C3-C5 Convertases/metabolism , Complement Membrane Attack Complex/analysis , Female , Follow-Up Studies , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Phenotype , Properdin/metabolism , Serologic Tests , Young Adult
12.
Kidney Int ; 92(4): 876-887, 2017 10.
Article in English | MEDLINE | ID: mdl-28729035

ABSTRACT

The intrinsic similarity shared between the members of the complement factor H family, which comprises complement factor H and five complement factor H-related (CFHR) genes, leads to various recombination events. In turn these events lead to deletions of some genes or abnormal proteins, which are found in patients with atypical hemolytic uremic syndrome or C3 glomerulopathies. Here we describe a novel genetic rearrangement generated from a heterozygous deletion spanning 146 Kbp involving multiple CFHR genes leading to a CFHR1-R5 hybrid protein. This deletion was found in four family members presenting with a familial dominant glomerulopathy histologically classified as an overlap of dense deposit disease and C3 glomerulonephritis. Affected patients exhibited permanently low C3 and factor B levels and high amounts of activation fragments sC5b9 and Bb, indicating a systemic alternative pathway dysregulation. The abnormal protein, characterized by Western blot and immunoprecipitation, was shown to circulate in association with CFHR1 and CFHR2, attributable to its two N-terminal dimerization motifs. The presence of this protein is associated with a perturbation of Factor H activity on the C3 convertase decay. Thus, our study highlights the role of CFHRs in the physiopathology of C3 glomerulopathies and stresses the importance of screening CFHRs in all familial C3 glomerulopathies. Such hybrids described till now were always associated with familial forms.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Complement C3/analysis , Complement C3b Inactivator Proteins/genetics , Complement System Proteins/genetics , Glomerulonephritis, Membranoproliferative/genetics , Adult , Atypical Hemolytic Uremic Syndrome/blood , Atypical Hemolytic Uremic Syndrome/pathology , Atypical Hemolytic Uremic Syndrome/therapy , Child , Complement C3-C5 Convertases/metabolism , Complement Factor B/analysis , Complement Factor H/metabolism , Complement Pathway, Alternative/genetics , Female , Gene Fusion , Gene Rearrangement , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/pathology , Glomerulonephritis, Membranoproliferative/therapy , Humans , Infant , Kidney/pathology , Male , Pedigree , Sequence Deletion
13.
N Engl J Med ; 371(4): 303-12, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25054716

ABSTRACT

BACKGROUND: Although thrombosis is considered the cardinal feature of the antiphospholipid syndrome, chronic vascular lesions are common, particularly in patients with life-threatening complications. In patients who require transplantation, vascular lesions often recur. The molecular pathways involved in the vasculopathy of the antiphospholipid syndrome are unknown, and adequate therapies are lacking. METHODS: We used double immunostaining to evaluate pathway activation in the mammalian target of rapamycin complex (mTORC) and the nature of cell proliferation in the vessels of patients with primary or secondary antiphospholipid syndrome nephropathy. We also evaluated autopsy specimens from persons who had catastrophic antiphospholipid syndrome. The molecular pathways through which antiphospholipid antibodies modulate the mTORC pathway were evaluated in vitro, and potential pharmacologic inhibitors were also tested in vitro. Finally, we studied the effect of sirolimus in kidney-transplant recipients with the antiphospholipid syndrome. RESULTS: The vascular endothelium of proliferating intrarenal vessels from patients with antiphospholipid syndrome nephropathy showed indications of activation of the mTORC pathway. In cultured vascular endothelial cells, IgG antibodies from patients with the antiphospholipid syndrome stimulated mTORC through the phosphatidylinositol 3-kinase (PI3K)-AKT pathway. Patients with antiphospholipid syndrome nephropathy who required transplantation and were receiving sirolimus had no recurrence of vascular lesions and had decreased vascular proliferation on biopsy as compared with patients with antiphospholipid antibodies who were not receiving sirolimus. Among 10 patients treated with sirolimus, 7 (70%) had a functioning renal allograft 144 months after transplantation versus 3 of 27 untreated patients (11%). Activation of mTORC was also found in the vessels of autopsy specimens from patients with catastrophic antiphospholipid syndrome. CONCLUSIONS: Our results suggest that the mTORC pathway is involved in the vascular lesions associated with the antiphospholipid syndrome. (Funded by INSERM and others.).


Subject(s)
Antibodies, Antiphospholipid/metabolism , Antiphospholipid Syndrome/metabolism , Endothelium, Vascular/metabolism , Immunosuppressive Agents/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adult , Analysis of Variance , Antiphospholipid Syndrome/complications , Antiphospholipid Syndrome/drug therapy , Autopsy , Cell Proliferation , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Female , Humans , Immunoglobulin G , Immunosuppressive Agents/therapeutic use , Kidney/blood supply , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Transplantation , Male , Metabolic Networks and Pathways/drug effects , Middle Aged , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism
14.
J Immunol ; 194(11): 5129-38, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25917093

ABSTRACT

Autoantibodies targeting factor H (FH), which is a main alternative complement pathway regulatory protein, have been well characterized in atypical hemolytic uremic syndrome (aHUS) but have been less well described in association with alternative pathway-mediated glomerulopathies (GP). In this study, we studied 17 patients presenting with GP who were positive for anti-FH IgG. Clinical data were collected and biological characteristics were compared with those of patients presenting with anti-FH Ab-associated aHUS. In contrast to the aHUS patients, the GP patients had no circulating FH-containing immune complexes, and their anti-FH IgG had a weaker affinity for FH. Functional studies demonstrated that these Abs induced no perturbations in FH cell surface protection or the binding of FH to its ligand. However, anti-FH IgG samples isolated from three patients were able to affect the factor I cofactor activity of FH. Epitope mapping identified the N-terminal domain of FH as the major binding site for GP patient IgG. No homozygous deletions of the CFHR1 and CFHR3 genes, which are frequently associated with the anti-FH Ab in aHUS patients, were found in the GP patients. Finally, anti-FH Abs were frequently associated with the presence of C3 nephritic factor in child GP patients and with monoclonal gammopathy in adult GP patients, who frequently showed Ig Lchain restriction during reactivity against factor H. These data provide deeper insights into the pathophysiological differences between aHUS and GP, demonstrating heterogeneity of anti-FH IgG.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Autoantibodies/immunology , Complement Factor H/antagonists & inhibitors , Glomerulonephritis, Membranoproliferative/immunology , Adolescent , Adult , Aged , Binding Sites , Blood Proteins/genetics , Child , Complement C3b Inactivator Proteins/genetics , Complement Factor H/immunology , Complement Factor I/immunology , Epitope Mapping , Female , Humans , Immunoglobulin G/immunology , Kidney Diseases/immunology , Male , Middle Aged , Young Adult
17.
J Biol Chem ; 290(42): 25343-55, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26245903

ABSTRACT

Lupus nephritis (LN) is a complication of the autoimmune disease systemic lupus erythematosus. Because the complement system plays a critical role in orchestrating inflammatory and immune responses as well as in the clearance of immune complexes, autoreactivity to complement components may have considerable pathological consequences. Autoantibodies against the central complement component C3 have been reported in systemic lupus erythematosus, but their molecular mechanism and functional relevance are not well understood. The objective of this study was to evaluate the frequency and the functional properties of the anti-C3 autoantibodies. Anti-C3 autoantibodies were measured in plasma of 39 LN patients, and identification of their epitopes on the C3 molecule was performed. By using surface plasmon resonance, we analyzed the influence of patient-derived IgG antibodies on the interaction of C3b with Factor B, Factor H, and complement receptor 1. The capacity of these antibodies to dysregulate the C3 convertase on the surface of endothelial cell was measured by flow cytometry. Here we report that the frequency of anti-C3 autoantibodies in LN is ∼30%. They inhibited interactions of the negative complement regulators Factor H and complement receptor 1 with C3b. An enhanced C3 deposition was also observed on human endothelial cells in the presence of C3 autoantibodies. In addition, anti-C3 autoantibody levels correlated with disease activity. In conclusion, the anti-C3 autoantibodies in LN may contribute to the autoimmune pathology by their capacity to overactivate the complement system.


Subject(s)
Autoantibodies/immunology , Complement C3/immunology , Lupus Nephritis/immunology , Adult , Aged , Cohort Studies , Female , Humans , Male , Middle Aged , Young Adult
20.
J Immunol ; 191(2): 912-21, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772024

ABSTRACT

Complement is an essential humoral component of innate immunity; however, its inappropriate activation leads to pathology. Polymorphisms, mutations, and autoantibodies affecting factor H (FH), a major regulator of the alternative complement pathway, are associated with various diseases, including age-related macular degeneration, atypical hemolytic uremic syndrome, and C3 glomerulopathies. Restoring FH function could be a treatment option for such pathologies. In this article, we report on an engineered FH construct that directly combines the two major functional regions of FH: the N-terminal complement regulatory domains and the C-terminal surface-recognition domains. This minimal-size FH (mini-FH) binds C3b and has complement regulatory functions similar to those of the full-length protein. In addition, we demonstrate that mini-FH binds to the FH ligands C-reactive protein, pentraxin 3, and malondialdehyde epitopes. Mini-FH was functionally active when bound to the extracellular matrix and endothelial cells in vitro, and it inhibited C3 deposition on the cells. Furthermore, mini-FH efficiently inhibited complement-mediated lysis of host-like cells caused by a disease-associated FH mutation or by anti-FH autoantibodies. Therefore, mini-FH could potentially be used as a complement inhibitor targeting host surfaces, as well as to replace compromised FH in diseases associated with FH dysfunction.


Subject(s)
Complement C3b/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Amino Acid Sequence , Base Sequence , C-Reactive Protein/metabolism , Complement Activation , Complement C3b/antagonists & inhibitors , Complement C3b/metabolism , Complement Pathway, Alternative , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Humans , Malondialdehyde/metabolism , Protein Binding , Protein Engineering , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Serum Amyloid P-Component/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL