Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Front Immunol ; 12: 654652, 2021.
Article in English | MEDLINE | ID: mdl-34276649

ABSTRACT

Objectives: This study aimed to determine the prevalence and localization of complement factor C4d in renal biopsies from patients with lupus nephritis (LN), as well as its associations with the disease's clinico-pathological features. The correlation between arteriolar C4d deposition and renal microvascular lesions (RVLs) was further analyzed. Methods: A total of 325 biopsy-proven LN patients were enrolled, and their clinico-pathological data were collected. C4d staining of renal biopsies was performed by immunohistochemistry. The associations between C4d deposition and the clinico-pathological features were further analyzed. Results: C4d deposition was present in most (98.8%) renal specimens in our cohort. These deposits were localized in the glomeruli (98.2%), tubular basement membrane (TBM) (43.7%), arterioles (31.4%), and peritubular capillary (33.8%). Patients with TBM C4d staining had higher disease activity (measured with the Systemic Lupus Erythematous Disease Activity Index) and higher National Institutes of Health pathological activity and chronicity indices (all P < 0.01). Patients with arteriolar C4d deposition were more likely to develop RVLs (91.2%) compared to those with no arteriolar C4d deposition (78.0%; P = 0.004), especially with two or more types of RVLs (P < 0.001). During the mean follow-up of 55.8 months, arteriolar C4d was related to worse renal outcomes [hazard ration (HR): 2.074, 95% confidence interval (CI) 1.056-4.075, P = 0.034]. Multivariate Cox hazard analysis showed that co-deposition of arteriolar C4d and C3c was an independent risk factor (HR: 3.681, 95% CI 1.519-8.921, P = 0.004) for predicting renal outcomes. Conclusions: C4d deposition was common in renal tissues from LN patients. TBM C4d deposition was related to the disease activity, and arteriolar C4d deposition was associated with RVLs and worse renal outcomes.


Subject(s)
Complement C4b/immunology , Complement C4b/metabolism , Disease Susceptibility , Lupus Nephritis/etiology , Lupus Nephritis/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Adult , Biomarkers , Biopsy , Complement C1q/immunology , Complement C1q/metabolism , Complement C3c/immunology , Complement C3c/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Kidney Glomerulus/immunology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lupus Nephritis/diagnosis , Lupus Nephritis/mortality , Male , Middle Aged , Prognosis , Young Adult
2.
J Infect Dis ; 223(2): 214-224, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33038254

ABSTRACT

BACKGROUND: Excessive activation of immune responses in coronavirus disease 2019 (COVID-19) is considered to be related to disease severity, complications, and mortality rate. The complement system is an important component of innate immunity and can stimulate inflammation, but its role in COVID-19 is unknown. METHODS: A prospective, longitudinal, single center study was performed in hospitalized patients with COVID-19. Plasma concentrations of complement factors C3a, C3c, and terminal complement complex (TCC) were assessed at baseline and during hospital admission. In parallel, routine laboratory and clinical parameters were collected from medical files and analyzed. RESULTS: Complement factors C3a, C3c, and TCC were significantly increased in plasma of patients with COVID-19 compared with healthy controls (P < .05). These complement factors were especially elevated in intensive care unit patients during the entire disease course (P < .005 for C3a and TCC). More intense complement activation was observed in patients who died and in those with thromboembolic events. CONCLUSIONS: Patients with COVID-19 demonstrate activation of the complement system, which is related to disease severity. This pathway may be involved in the dysregulated proinflammatory response associated with increased mortality rate and thromboembolic complications. Components of the complement system might have potential as prognostic markers for disease severity and as therapeutic targets in COVID-19.


Subject(s)
COVID-19/immunology , Complement Activation/immunology , SARS-CoV-2/immunology , Aged , Aged, 80 and over , COVID-19/mortality , Complement C3c/immunology , Cytokines/blood , Disease Progression , Female , Humans , Immunity, Innate , Inflammation/immunology , Longitudinal Studies , Male , Middle Aged , Mortality , Netherlands/epidemiology , Prospective Studies , Respiratory Distress Syndrome/immunology , Severity of Illness Index
3.
Clin Immunol ; 221: 108616, 2020 12.
Article in English | MEDLINE | ID: mdl-33148511

ABSTRACT

In complement-driven thrombotic microangiopathies, failure to regulate complement activation leads to end-organ damage. The modified Ham (mHam) test measures complement-mediated killing of a nucleated cell in vitro but lacks a confirmatory assay and reliable positive controls. We demonstrate that C5b-9 accumulation on the surface of TF1 PIGAnull cells correlates with cell killing in the mHam. We also show that Sialidase treatment of cells or addition of Shiga toxin 1 to human serum serve as a more reliable positive control for the mHam than cobra venom factor or lipopolysaccharide. Simultaneously performing the mHam and measuring C5b-9 accumulation either in GVB++ or GVB0 MgEGTA buffer with the addition of complement pathway specific inhibitors (anti-C5 antibody or a factor D inhibitor, ACH-145951) can be used to localize defects in complement regulation. As more targeted complement inhibitors become available, these assays may aid in the selection of personalized treatments for patients with complement-mediated diseases.


Subject(s)
Antiphospholipid Syndrome/immunology , Atypical Hemolytic Uremic Syndrome/immunology , Complement Activation/drug effects , Complement Inactivating Agents/pharmacology , Adult , Biological Assay , Cell Line, Tumor , Complement C3c/immunology , Complement C4b/immunology , Complement Membrane Attack Complex/immunology , Elapid Venoms/pharmacology , Female , Humans , Lipopolysaccharides/pharmacology , Male , Middle Aged , Neuraminidase/pharmacology , Peptide Fragments/immunology , Shiga Toxin 1/pharmacology
4.
Blood ; 132(23): 2431-2440, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30309891

ABSTRACT

The mechanisms by which exposure to heparin initiates antibody responses in many, if not most, recipients are poorly understood. We recently demonstrated that antigenic platelet factor 4 (PF4)/heparin complexes activate complement in plasma and bind to B cells. Here, we describe how this process is initiated. We observed wide stable variation in complement activation when PF4/heparin was added to plasma of healthy donors, indicating a responder "phenotype" (high, intermediate, or low). Proteomic analysis of plasma from these healthy donors showed a strong correlation between complement activation and plasma immunoglobulin M (IgM) levels (r = 0.898; P < .005), but not other Ig isotypes. Complement activation response to PF4/heparin in plasma displaying the low donor phenotype was enhanced by adding pooled IgM from healthy donors, but not monoclonal IgM. Depletion of IgM from plasma abrogated C3c generation by PF4/heparin. The complement-activating features of IgM are likely mediated by nonimmune, or natural, IgM, as cord blood and a monoclonal polyreactive IgM generate C3c in the presence of PF4/heparin. IgM facilitates complement and antigen deposition on B cells in vitro and in patients receiving heparin. Anti-C1q antibody prevents IgM-mediated complement activation by PF4/heparin complexes, indicating classical pathway involvement. These studies demonstrate that variability in plasma IgM levels correlates with functional complement responses to PF4/heparin. Polyreactive IgM binds PF4/heparin, triggers activation of the classical complement pathway, and promotes antigen and complement deposition on B cells. These studies provide new insights into the evolution of the heparin-induced thrombocytopenia immune response and may provide a biomarker of risk.


Subject(s)
B-Lymphocytes/immunology , Complement Pathway, Classical/immunology , Heparin/immunology , Immunoglobulin M/immunology , Lymphocyte Activation , Platelet Factor 4/immunology , Complement C3c/immunology , Complement Pathway, Classical/drug effects , Heparin/pharmacology , Humans , Platelet Factor 4/pharmacology , Proteomics
5.
Clin Exp Immunol ; 189(3): 372-382, 2017 09.
Article in English | MEDLINE | ID: mdl-28509333

ABSTRACT

The goal of this study was to investigate the glycosylation profile of native immunoglobulin (Ig)G present in serum immune complexes in patients with rheumatoid arthritis (RA). To accomplish this, lectin binding assays, detecting the accessibility of glycans present on IgG-containing immune complexes by biotinylated lectins, were employed. Lectins capturing fucosyl residues (AAL), fucosylated tri-mannose N-glycan core sites (LCA), terminal sialic acid residues (SNA) and O-glycosidically linked galactose/N-acetylgalactosamine (GalNac-L) were used. Patients with recent-onset RA at baseline and after 3-year follow-up were investigated. We found that native IgG was complexed significantly more often with IgM, C1q, C3c and C-reactive protein (CRP) in RA patients, suggesting alterations of the native structure of IgG. The total accessibility of fucose residues on captured immune complexes to the respective lectin was significantly higher in patients with RA. Moreover, fucose accessibility on IgG-containing immune complexes correlated positively with the levels of antibodies to cyclic citrullinated peptides (anti-CCP). We also observed a significantly higher accessibility to sialic acid residues and galactose/GalNAc glyco-epitopes in native complexed IgG of patients with RA at baseline. While sialic acid accessibility increased during treatment, the accessibility of galactose/GalNAc decreased. Hence, successful treatment of RA was associated with an increase in the SNA/GalNAc-L ratio. Interestingly, the SNA/GalNAc-L ratio in particular rises after glucocorticoid treatment. In summary, this study shows the exposure of glycans in native complexed IgG of patients with early RA, revealing particular glycosylation patterns and its changes following pharmaceutical treatment.


Subject(s)
Antigen-Antibody Complex/immunology , Arthritis, Rheumatoid/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Polysaccharides/chemistry , Polysaccharides/immunology , Adult , Aged , Antigen-Antibody Complex/chemistry , Arthritis, Rheumatoid/therapy , C-Reactive Protein/immunology , C-Reactive Protein/metabolism , Complement C1q/immunology , Complement C1q/metabolism , Complement C3c/immunology , Complement C3c/metabolism , Female , Fucose/metabolism , Galactose/metabolism , Glycosylation , Humans , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Lectins/metabolism , Male , Middle Aged , Polysaccharides/metabolism , Sambucus nigra , Sialic Acids/metabolism
6.
J Immunol Res ; 2015: 638129, 2015.
Article in English | MEDLINE | ID: mdl-26185769

ABSTRACT

To analyze the glycosylation of anti-ß2GP1, we investigated purified IgG from healthy children, patients with APS, and asymptomatic adult carriers of antiphospholipid antibodies. We observed that in the sera of healthy children and of patients with APS, IgG3 and IgG2 were predominant, respectively. The potentially protective anti-ß2GP1-IgM was lower in the sera of healthy children. Although anti-ß2GP1-associated C1q did not differ between children and patients with antiphospholipid syndrome, the associated C3c was significantly higher in the sera of healthy children. This indicates a more efficient clearance of anti-ß2GP1 immune complexes in the healthy children. This clearance is not accompanied by inflammation or coagulatory events. It is likely that the most important pathogenic factor of the anti-ß2GP1-IgG is related to the different glycosylation observed in healthy and diseased individuals. We detected a significantly higher sialylation of anti-ß2GP1-IgG isolated from the sera of healthy children and asymptomatic adults when compared with that of patients with clinically apparent antiphospholipid syndrome. Low sialylated IgG reportedly ameliorates inflammation and inflammation promotes hyposialylation. Thus, both reactions create a vicious circle that precipitates the pathology of the antiphospholipid syndrome including thrombus-formation. We conclude that the increased sialylation of anti-ß2GP1-IgG of sera of healthy individuals limits their pathogenicity.


Subject(s)
Autoantibodies/immunology , Autoantibodies/metabolism , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , beta 2-Glycoprotein I/immunology , Adolescent , Antibodies, Antiphospholipid/immunology , Antiphospholipid Syndrome/immunology , Antiphospholipid Syndrome/metabolism , Case-Control Studies , Child , Child, Preschool , Complement C1q/immunology , Complement C1q/metabolism , Complement C3c/immunology , Complement C3c/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Glycosylation , Humans , Immunoglobulin M/immunology , Infant , Infant, Newborn , Male , beta 2-Glycoprotein I/antagonists & inhibitors
7.
J Immunol ; 195(1): 257-64, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26026058

ABSTRACT

Chronic inflammation of the arterial wall is a key element in the development of atherosclerosis, and cholesterol crystals (CC) that accumulate in plaques are associated with initiation and progression of the disease. We recently revealed a link between the complement system and CC-induced inflammasome caspase-1 activation, showing that the complement system is a key trigger in CC-induced inflammation. HDL exhibits cardioprotective and anti-inflammatory properties thought to explain its inverse correlation to cardiovascular risk. In this study, we sought to determine the effect of reconstituted HDL (rHDL) on CC-induced inflammation in a human whole blood model. rHDL bound to CC and inhibited the CC-induced complement activation as measured by soluble terminal C5b-9 formation and C3c deposition on the CC surface. rHDL attenuated the amount of CC-induced complement receptor 3 (CD11b/CD18) expression on monocytes and granulocytes, as well as reactive oxygen species generation. Moreover, addition of CC to whole blood resulted in release of proinflammatory cytokines that were inhibited by rHDL. Our results support and extend the notion that CC are potent triggers of inflammation, and that rHDL may have a beneficial role in controlling the CC-induced inflammatory responses by inhibiting complement deposition on the crystals.


Subject(s)
Cholesterol/adverse effects , Complement Activation/drug effects , Lipoproteins, HDL/pharmacology , Blood Cells/cytology , Blood Cells/drug effects , Blood Cells/immunology , CD11b Antigen/immunology , CD18 Antigens/immunology , Complement C3c/antagonists & inhibitors , Complement C3c/immunology , Complement Membrane Attack Complex/antagonists & inhibitors , Complement Membrane Attack Complex/immunology , Crystallization , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Primary Cell Culture , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/immunology
8.
J Immunol Methods ; 419: 25-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25733354

ABSTRACT

The complement system is an essential component of our innate immunity, both for the protection against infections and for proper handling of dying cells. However, the complement system can also contribute to tissue injury and inflammatory responses. In view of novel therapeutic possibilities, there is an increasing interest in measurement of the complement system activation in the systemic compartment, both in the clinical setting as well as in experimental models. Here we describe in parallel a sensitive and specific sandwich ELISA detecting mouse C3 activation fragments C3b/C3c/iC3b, as well as functional complement ELISAs detecting specific activities of the three complement pathways at the level of C3 and at the level of C9 activation. In a murine model of renal ischaemia/reperfusion injury (IRI) we found transient complement activation as shown by generation of C3b/C3c/iC3b fragments at 24 h following reperfusion, which returned to base-line at 3 and 7 days post reperfusion. When the pathway specific complement activities were measured at the level of C3 activation, we found no significant reduction in any of the pathways. However, the functional complement activity of all three pathways was significantly reduced when measured at the level of C9, with the strongest reduction being observed in the alternative pathway. For all three pathways there was a strong correlation between the amount of C3 fragments and the reduction in functional complement activity. Moreover, at 24 h both C3 fragments and the functional complement activities showed a correlation with the rise in serum creatinine. Together our results show that determination of the systemic pathway specific complement activity is feasible in experimental mouse models and that they are useful in understanding complement activation and inhibition in vivo.


Subject(s)
Complement Activation/immunology , Complement C3b/immunology , Complement C3c/immunology , Kidney/immunology , Reperfusion Injury/immunology , Animals , Complement Activation/genetics , Complement C3b/genetics , Complement C3b/metabolism , Complement C3c/genetics , Complement C3c/metabolism , Complement C9/immunology , Complement C9/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Feasibility Studies , Kidney/blood supply , Kidney/metabolism , Mice, Inbred C57BL , Mice, Knockout , Reproducibility of Results , Time Factors
9.
Clin Exp Immunol ; 181(1): 118-25, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25677399

ABSTRACT

Haemolytic uraemic syndrome (HUS) is characterized by haemolytic anaemia, thrombocytopenia and acute renal failure. We studied the activation state of classical and alternative pathways of complement during the acute phase of Shiga toxin-associated HUS by performing a prospective study of 18 patients and 17 age-matched healthy controls to evaluate C3, C3c, C4, C4d, Bb and SC5b-9 levels. SC5b-9 levels were increased significantly in all patients at admission compared to healthy and end-stage renal disease controls, but were significantly higher in patients presenting with oliguria compared to those with preserved diuresis. C3 and C4 levels were elevated significantly at admission in the non-oliguric group when compared to controls. No significant differences were found for C4d values, whereas factor Bb was elevated in all patients and significantly higher in oliguric patients when compared to both controls and non-oliguric individuals. A positive and significant association was detected when Bb formation was plotted as a function of plasma SC5b-9 at admission. Bb levels declined rapidly during the first week, with values not significantly different from controls by days 3 and 5 for non-oligurics and oligurics, respectively. Our data demonstrate the activation of the alternative pathway of complement during the acute phase of Stx-associated HUS. This finding suggests that complement activation may represent an important trigger for the cell damage that occurs during the syndrome.


Subject(s)
Complement Activation/immunology , Complement C3-C5 Convertases, Alternative Pathway/immunology , Complement Membrane Attack Complex/immunology , Hemolytic-Uremic Syndrome/immunology , Adolescent , Adult , Child , Complement C3/immunology , Complement C3c/immunology , Complement C4/immunology , Complement C4b/immunology , Female , Humans , Kidney/immunology , Kidney/pathology , Male , Middle Aged , Peptide Fragments/blood , Peptide Fragments/immunology , Prospective Studies , Shiga Toxin/immunology , Young Adult
10.
Am J Trop Med Hyg ; 88(2): 230-5, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23390222

ABSTRACT

Angiostrongylus cantonensis is a zoonotic pathogen that occasionally causes human angiostrongyliasis; its main clinical manifestation is eosinophilic meningitis. This report defines the concept of intrathecal activation of complement as evidence of intrathecal synthesis of major immunoglobulins during this disease. Details are presented of the activation of complement system components in cerebrospinal fluid, and their application to our understanding of this tropical disease, which is emerging in the Western hemisphere. Intrathecal synthesis of at least one of the major immunoglobulins and a wide spectrum of patterns may be observed. Although intrathecal synthesis of C3c is always present, C4 intrathecal synthesis does not occur in every patient. The diversity of intrathecal synthesis and activation of the different complement pathways enables their division into three variant groups (A, B, and C). Variant group A includes the classical and/or lectin pathway and involves two or more major immunoglobulins with C3 and C4 intrathecal synthesis. Variant group B involves C4 in cerebrospinal fluid that comes from blood in the intrathecal activation of the classical pathway. Variant group C includes the alternative pathway.


Subject(s)
Central Nervous System/immunology , Central Nervous System/parasitology , Strongylida Infections/immunology , Angiostrongylus cantonensis/isolation & purification , Angiostrongylus cantonensis/pathogenicity , Animals , Complement C3c/cerebrospinal fluid , Complement C3c/immunology , Complement C4b/cerebrospinal fluid , Complement C4b/immunology , Eosinophilia/cerebrospinal fluid , Eosinophilia/immunology , Eosinophilia/parasitology , Humans , Immunoglobulins/cerebrospinal fluid , Immunoglobulins/immunology , Meningitis/cerebrospinal fluid , Meningitis/immunology , Meningitis/parasitology , Peptide Fragments/cerebrospinal fluid , Peptide Fragments/immunology , Strongylida Infections/cerebrospinal fluid , Strongylida Infections/parasitology
11.
Autoimmunity ; 46(3): 205-14, 2013 May.
Article in English | MEDLINE | ID: mdl-23331132

ABSTRACT

The pattern recognition molecules C-reactive protein (CRP) and C1q are of big interest in relation to the pathogenesis of systemic lupus erythematosus (SLE). Circulating autoantibodies against CRP and C1q are frequently found in SLE patients with active disease, particularly in lupus nephritis (LN), and rising levels reportedly relate to disease activity and outcome. If CRP-, or dsDNA- and/or C1q-containing immune complexes (ICs) are pathogenic in LN, glomerular IgG-deposits would be expected to co-localize with these antigens. In search for proof of this concept, renal biospsies from patients with active LN (n = 5) were examined with high-resolution immunogold electron microscopy. Renal biopsies from patients with Henoch-Schönlein purpura, pauci-immune nephritis and renal cancer served as controls. IgG antibodies against CRP, C1q and nucleosomes were analyzed in pre-post flare sera. We could demonstrate that CRP, C1q, C3c and dsDNA were co-localized with IgG in electron dense deposits in the glomerular basement membrane/subendothelial space in all of the 5 LN patients. Deposits of IgG, CRP, complement and dsDNA were 10-fold higher in LN compared to controls. All SLE patients had circulating anti-nucleosome antibodies; 4/5 had serum antibodies against CRP, dsDNA, and C1q at biopsy/flare. Despite a limited number of cases, the results support the notion of a pathogenic role not only for anti-dsDNA antibodies, but also for anti-CRP and anti-C1q in LN. The glomerular ICs may have been generated by deposition of circulating ICs or by in situ IC formation.


Subject(s)
Antibodies, Antinuclear/immunology , Autoantibodies/blood , C-Reactive Protein/immunology , Complement C1q/immunology , Lupus Nephritis/immunology , Adult , Aged , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Antibodies, Antinuclear/blood , Antigen-Antibody Complex/blood , C-Reactive Protein/metabolism , Complement C1q/metabolism , Complement C3c/immunology , DNA/immunology , Female , Humans , IgA Vasculitis/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Kidney/pathology , Kidney Neoplasms/immunology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Male , Middle Aged , Nephritis/immunology , Nucleosomes/immunology
12.
Tissue Antigens ; 80(5): 404-15, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22931381

ABSTRACT

Donor human leukocyte antigen (HLA)-specific antibodies (Abs) with the ability to activate complement are associated with an increased risk of early Ab-mediated rejection (AMR) of kidney allografts. In recent years, also non-HLA Abs-binding endothelial cells have been shown to elicit early AMR. Donor-specific anti-endothelial cell Abs escape detection in the pre-transplant evaluation if only lymphocytes are used as target cells in crossmatch tests. We addressed whether endothelial precursor cells (EPCs) could be used for detection of complement-fixing as well as non-fixing Abs and if complement factor and immunoglobulin G (IgG) deposition on co-purified T and B cells correlated to the outcome of the T- and B-cell complement-dependent cytotoxicity assay. Deposition of complement factors C3c and C3d, but not C1q nor C4d, were detected on EPCs and lymphocytes upon incubation with HLA Ab-positive sera. There was a correlation between the amount of C3c deposition and IgG binding on EPCs (R(2) = 0.71, P = 0.0012) and T cells (R(2) = 0.74, P = 0.0006) but not for B cells (R(2) = 0.34, P = 0.059). The specificity and sensitivity for C3d deposition on endothelial precursor cell crossmatch (EPCXM) T cells vs the T complement-dependent cytotoxicity (CDC) assay were 69% and 72%, respectively. The EPCXM B-cell C3d assay had considerably lower sensitivity (39%) than the B CDC assay. Altogether, this novel assay based on the detection of complements factors on EPCs and lymphocytes by flow cytometry may widen the diagnostic repertoire and thereby improve the clinical management of patients undergoing kidney transplantation.


Subject(s)
B-Lymphocytes/immunology , Endothelial Cells/immunology , Flow Cytometry/methods , Graft Rejection/immunology , Isoantibodies/analysis , Kidney Transplantation/immunology , T-Lymphocytes/immunology , Antibody Specificity , B-Lymphocytes/chemistry , Cell Differentiation , Complement C1q/immunology , Complement C3c/immunology , Complement C3d/immunology , Complement C4b/immunology , Cytotoxicity Tests, Immunologic , Endothelial Cells/chemistry , Graft Rejection/diagnosis , HLA Antigens/immunology , Humans , Immunoglobulin G/immunology , Isoantibodies/immunology , Male , Peptide Fragments/immunology , Sensitivity and Specificity , T-Lymphocytes/chemistry , Transplantation, Homologous
13.
Histopathology ; 57(6): 825-35, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21166697

ABSTRACT

AIMS: Autoimmune pancreatitis (AIP) is a type of pancreatitis whose immunopathogenesis is still unknown. It has been reported that renal biopsy specimens from patients diagnosed with both AIP and tubulointerstitial nephritis reveal deposits containing complement C3, immunoglobulin (Ig)G and IgG4 at the tubular basement membranes (BMs). The aim was to investigate the deposition of complement and immunoglobulins in pancreatic tissue from AIP patients compared to non-AIP patients. METHODS: Double immunofluorescence microscopy for C3c, IgG4 and IgG together with CK7, trypsin, collagen IV, CD31 and CD79a, as well as immunofluorescence microscopy for C1q, IgA and IgM, were performed on frozen pancreatic tissue from AIP and alcoholic chronic pancreatitis (ACP) patients. RESULTS: In AIP patients, complement C3c, IgG4 and IgG were deposited at the collagen IV-positive BMs of pancreatic and bile ducts and of acini. In a minority of the ACP patients, weak C3c-positive BM deposits were detected, but no IgG4- or IgG-positive BM deposits were present. CONCLUSION: The deposition of C3c, IgG4 and IgG at the BM of small- and medium-sized ducts and acini of the pancreas is characteristic of AIP. This suggests that immune complex-mediated destruction of ducts and acini play a role in the pathogenesis of AIP.


Subject(s)
Autoimmune Diseases/pathology , Basement Membrane/pathology , Complement C3c/immunology , Immunoglobulin G/immunology , Pancreatic Ducts/pathology , Pancreatitis, Chronic/pathology , Aged , Autoimmune Diseases/immunology , Basement Membrane/immunology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Pancreatic Ducts/immunology , Pancreatitis, Alcoholic/immunology , Pancreatitis, Alcoholic/pathology , Pancreatitis, Chronic/immunology
14.
J Immunol Methods ; 362(1-2): 142-50, 2010 Oct 31.
Article in English | MEDLINE | ID: mdl-20869965

ABSTRACT

There is a general need for markers of systemic inflammation in acute or chronic diseases, where complement activation is involved. Available methods to monitor complement activation are elaborate and of low sensitivity; they include haemolytic assays (CH50), quantification of fluid phase terminal complex (C5b-C9) and quantification of complement split products by precipitation-in-gel techniques (e.g. C3d). We have developed a mouse monoclonal antibody (mAb) that is able to detect fluid phase C3c without interference from other products generated from the complement component C3. The C3c specific mAb was tested in different ELISA combinations with various types of in vitro activated sera and with plasma or serum samples from factor I deficient patients. The specificity of the mAb was evaluated in immunoprecipitation techniques and by analysis of eluted fragments of C3 after immunoaffinity chromatography. The C3c mAb was confirmed to be C3c specific, as it showed no cross-reactivity with native (un-cleaved) C3, with C3b, iC3b, or with C3d. Also, no significant reaction was observed with C3 fragments in factor I deficient sera or plasma. This antibody forms the basis for the generation of a robust ELISA that allows for a quick and reliable evaluation of complement activation and consumption as a marker for inflammatory processes. We established the C3c plasma range in 100 healthy Danish blood donors with a mean of 3.47 µg/ml and a range of 2.12-4.92 µg/ml. We believe that such an antibody might be of potential value in the assessment of in vivo complement activity during the inflammatory processes.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity , Complement Activation/immunology , Complement C3c/immunology , Inflammation Mediators/immunology , Animals , Antibodies, Monoclonal/chemistry , Biomarkers/blood , Blood Donors , Complement C3c/metabolism , Denmark , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Inflammation/blood , Inflammation/immunology , Inflammation Mediators/blood , Male , Mice , Mice, Inbred BALB C , Sensitivity and Specificity
15.
Neurobiol Dis ; 26(3): 497-511, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17412602

ABSTRACT

We investigated the involvement of the complement cascade during epileptogenesis in a rat model of temporal lobe epilepsy (TLE), and in the chronic epileptic phase in both experimental as well as human TLE. Previous rat gene expression analysis using microarrays indicated prominent activation of the classical complement pathway which peaked at 1 week after SE in CA3 and entorhinal cortex. Increased expression of C1q, C3 and C4 was confirmed in CA3 tissue using quantitative PCR at 1 day, 1 week and 3-4 months after status epilepticus (SE). Upregulation of C1q and C3d protein expression was confirmed mainly to be present in microglia and in a few hippocampal neurons. In human TLE with hippocampal sclerosis, astroglial, microglial and neuronal (5/8 cases) expression of C1q, C3c and C3d was observed particularly within regions where neuronal cell loss occurs. The membrane attack protein complex (C5b-C9) was predominantly detected in activated microglial cells. The persistence of complement activation could contribute to a sustained inflammatory response and could destabilize neuronal networks involved.


Subject(s)
Complement System Proteins/immunology , Encephalitis/immunology , Epilepsy, Temporal Lobe/immunology , Gliosis/immunology , Hippocampus/immunology , Up-Regulation/immunology , Adolescent , Adult , Aged , Animals , Astrocytes/immunology , Astrocytes/metabolism , Complement C1q/genetics , Complement C1q/immunology , Complement C1q/metabolism , Complement C3c/genetics , Complement C3c/immunology , Complement C3c/metabolism , Complement C3d/genetics , Complement C3d/immunology , Complement C3d/metabolism , Complement C5b/genetics , Complement C5b/immunology , Complement C5b/metabolism , Complement System Proteins/genetics , Complement System Proteins/metabolism , Disease Models, Animal , Encephalitis/genetics , Encephalitis/physiopathology , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/physiopathology , Female , Gliosis/genetics , Gliosis/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Male , Microglia/immunology , Microglia/metabolism , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Status Epilepticus/genetics , Status Epilepticus/immunology , Status Epilepticus/physiopathology , Up-Regulation/genetics
16.
Mol Immunol ; 43(3): 236-45, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16199260

ABSTRACT

In periodontal disease, IgG1 and IgA1 antibodies produced in situ deposit on antigens in the affected tissues. Thus, there is an interest in the effect of co-deposited IgA1 antibodies on complement activation by IgG1-immune complexes. In the present study, we first analyzed the effect of IgA1-immune complexes on complement using human IgA1 antibodies to dansyl (with dansylated human serum albumin serving as the immobilized antigen). It was observed that these IgA1-immune complexes when incubated for prolonged times with 33% human serum as a source of complement received C4b and C3b deposition. As C4b and C3b deposited on the IgA1 antibodies and on the antigenic surface, the complement-coated IgA1 antibodies departed. These fluid-phase complement-coated IgA1 antibodies were transferred to antigen-coated microtiter-ELISA plates, where they became bound to the antigens. Thus, the complement-coated IgA1 antibodies retained their antigen-binding function, especially as a proportion of their covalently bound C3b progressively degraded to iC3b and C3d. Genetically engineered carbohydrate-deficient mutant human IgA1 antibodies were used to assess the role of carbohydrate in accepting the C4b and C3b depositions, and these studies indicated that the carbohydrate on the Fc-region of IgA1 played a positive role. Another interesting finding generated by this study was that when IgA1 was co-deposited with IgG1 antibodies, and serum complement was added, the IgG1 antibodies tended to remain on the antigenic surface. The co-deposited IgA1 antibodies not only controlled (reduced) the rate of the consumption of the first component of complement (C1) and of classical complement pathway activation by IgG1-immune complexes (and therein reduced the rate of complement-mediated dissolution of the IgG1-immune complexes), but also the co-deposited IgA1 antibodies simultaneously intercepted/accepted C4b and C3b, then departed, as complement began to cover the antigenic surfaces. The process in which complement-coated IgA1 antibodies transferred to non-complement-coated antigens is termed complement-coated antibody-transfer/transport (CCAT). In this way, IgA1 antibodies extended the efficiency of the complement system by insuring the specific IgA1 antibody-mediated transport of the captured biologically active complement fragments to those antigens stimulating the IgA1 antibody response but not yet neutralized (completely coated) with complement. Simultaneously by impeding the rate of C1 consumption and by intercepting C4b and C3b, IgA1 antibodies slowed C4b and C3b deposition on the antigenic surface and on the co-deposited IgG1 antibodies. Thus, in the presence of ongoing complement activation, the deposition of serum IgA1 antibodies enabled the co-deposited IgG1 antibodies to better maintain their ability to interact with antigens. We termed this latter phenomenon, preservation of IgG antibody deployment (PGD). In summary, co-deposited IgA1 antibodies maximized the efficiency of the complement system, transported their covalently bound complement fragments to specific antigens and sustained the effective deployment of IgG1 antibodies directed to those same antigens.


Subject(s)
Complement C3c/immunology , Complement C3d/immunology , Complement C4b/immunology , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Periodontal Diseases/immunology , Antigens/chemistry , Antigens/immunology , Biological Transport/immunology , Complement C3c/chemistry , Complement C3d/chemistry , Complement C4b/chemistry , Complement Pathway, Classical/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin A/chemistry , Immunoglobulin G/chemistry , Kinetics
17.
J Gastroenterol Hepatol ; 20(9): 1344-51, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16105119

ABSTRACT

BACKGROUND: It has previously been reported that there is a strong correlation between the expression of glycosylphosphatidylinositol (GPI)-anchored complement membrane inhibitor in gastric epithelium and the severity of inflammation of gastric mucosa. To investigate the regulation of complement activity in gastric epithelium during Helicobacter pylori (H. pylori)-associated gastritis, the expression of GPI-anchored complement membrane inhibitors, decay-accelerating factor (DAF) and 20-kDa homologous restriction factor 20 (HRF20), and membrane cofactor protein (MCP), which is a transmembrane protein, were evaluated after removal of the H. pylori stimulus. Furthermore, the expression of the complement fragment, C3c, was also investigated. METHODS: Forty-six patients with epigastric symptoms and endoscopically confirmed peptic ulcer or gastritis who had H. pylori infection of the gastric mucosa were enrolled in the present study. Biopsy specimens were obtained from the gastric antrum and corpus 1 month before and after eradication. Helicobacter pylori infection was determined by the rapid urease test, histology, and culture before eradication, and by histology, culture, and urea breath test after eradication. Gastric biopsy specimens obtained before and after eradication were evaluated for infiltration by neutrophils and mononuclear cells. The expression of complement membrane inhibitors, DAF, HRF20, and MCP and that of the main complement fragment, C3c, was immunohistochemically evaluated. RESULTS: One month after the eradication of H. pylori, the infiltration by neutrophils and mononuclear cells in the gastric mucosa decreased significantly (P < 0.0001) as compared with that before eradication. The expression of DAF, HRF20, and C3c on gastric mucosal epithelium also significantly decreased in both the antrum and the corpus (P < 0.05) 1 month after eradication. However, no change was observed in the expression of MCP. CONCLUSIONS: The decrease in the expression of GPI-anchored complement regulator and the complement after removal of a chronic microbial stimulus suggests that the gastric epithelium appears to undergo an aggressive stress of complement during H. pylori infection. Conclusively, DAF and HRF20 may play an important protective role against complement-mediated damage induced by chronic microbial stimuli in such a pathological condition.


Subject(s)
Complement System Proteins/immunology , Gastric Mucosa/immunology , Helicobacter Infections/immunology , Helicobacter pylori , Amoxicillin/therapeutic use , Anti-Bacterial Agents/therapeutic use , Anti-Ulcer Agents/therapeutic use , CD55 Antigens/biosynthesis , CD59 Antigens/biosynthesis , Clarithromycin/therapeutic use , Complement C3c/immunology , Complement Inactivator Proteins/biosynthesis , Female , Gastric Mucosa/physiopathology , Gastritis/drug therapy , Gastritis/immunology , Glycosylphosphatidylinositols/biosynthesis , Helicobacter Infections/drug therapy , Humans , Male , Membrane Cofactor Protein/biosynthesis , Middle Aged , Omeprazole/therapeutic use , Peptic Ulcer/drug therapy , Peptic Ulcer/immunology , Receptors, Complement/biosynthesis
18.
West Afr J Med ; 23(3): 253-5, 2004.
Article in English | MEDLINE | ID: mdl-15587841

ABSTRACT

OBJECTIVE: To evaluate serum levels of circulating immune complexes (CICs), immunoglobulin classes (IgG, IgA and IgM) and Complement Components (C3c, C4 and Factor B) in Nigerians with Type 1 or Type 2 diabetes mellitus. DESIGN: Case control study. SETTING: University College Hospital, Ibadan, Oyo State, Nigeria. SUBJECTS: Forty-two subjects with diabetic mellitus (17 Type 1 D. M. and 25 Type 2 D. M.) and 21 apparently healthy control subjects. INTERVENTION: Serum level of CICs was measured by polyethylene glycol precipitation method while single radial immunodiffusion method was used to measure serum levels of immunoglobulins and complement components. RESULTS: Only CICs were significantly higher in Type 1 diabetic subjects compared with the controls whereas CICs C3c, C4 and IgM were significantly increased in Type 2 diabetic subjects compared with the controls. The levels of CICs, C3c and IgM were significantly elevated in Type 2 diabetics compared with Type 1 diabetics. CONCLUSION: CIC concentrations may serve as a useful index of depressed host defences usually associated with diabetics mellitus and that humoral immunity is deranged more in Type 2 diabetics compared with Type 1 diabetics. Probably as a result of hyperinsulinaemia associated with insulin resistance.


Subject(s)
Antigen-Antibody Complex/blood , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/immunology , Adult , Aged , Case-Control Studies , Complement C3c/analysis , Complement C3c/immunology , Complement C4/analysis , Complement C4/immunology , Complement Factor B/analysis , Complement Factor B/immunology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Middle Aged , Nigeria
19.
Immunogenetics ; 55(12): 811-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14770272

ABSTRACT

The complement system helps in the lysis of invading pathogens and modulates the inflammatory as well as the humoral and cellular immune responses. C5 mediates many potent inflammatory and cytolytic events after proteolytic activation by complement convertase enzymes. Hence, to investigate the role of pig C5 ( pC5) as a candidate gene for disease resistance in pigs, the complete cDNA of pC5 was sequenced, screened for single nucleotide polymorphisms (SNPs), and an association analysis with various immunological parameters measured in F2 animals of a pig resource population based on a cross of Duroc and Berlin miniature pigs (DUMI) was carried out. In total, 5,422 bp of pC5 cDNA was sequenced, which codes for the 1,677-amino-acid precursor of C5. Four polymorphic sites were detected, one of which was segregating in the DUMI population in three genotypic patterns: AA, AC and CC. Classical (CH50) and alternative (AH50) complement activities, C3c levels, haptoglobin (HP) acute phase protein levels, and antibody titers against Mycoplasma (Mk) and Aujesky (ADV) vaccines were measured in the resource population. Association analysis between C5 and the immunological parameters was carried out using repeated measures mixed and general linear model analysis. The homozygote AA was found to be significantly different from the other two genotypes with respect to AH50 and CH50, whereas genotype CC was found to be significantly different from the other genotypes for C3c and HP levels. No significant difference could be seen between genotypes for antibody titers against vaccinations. Association of C5 with complement activity traits and acute phase proteins promotes pC5 as a candidate gene for innate disease resistance.


Subject(s)
Complement C5/genetics , Quantitative Trait Loci/immunology , Swine/immunology , Acute-Phase Proteins/metabolism , Animals , Base Sequence , Chromosome Segregation , Cloning, Molecular , Complement C3c/immunology , Complement Hemolytic Activity Assay , Complement Pathway, Alternative/immunology , Complement Pathway, Classical/immunology , DNA, Complementary/analysis , Female , Genotype , Haptoglobins/metabolism , Male , Molecular Sequence Data , Phenotype , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
20.
J Mol Med (Berl) ; 81(7): 404-10, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12827269

ABSTRACT

The complement system of components and receptors is one of the earliest forms of defence. Excessive or inappropriate activation can result in tissue damage, classically illustrated in immune-mediated nephritis. In addition, complement forms a bridge between innate and adaptive immunity, helping to prepare and focus T and B lymphocyte responses. More recent research in renal allograft models has shown that complement-inhibited and complement-deficient animals have reduced inflammatory injury and lowered antidonor immune responses. Furthermore, it is known that the transplanted kidney is a significant site of local synthesis of C3, although until recently the relative contribution of locally produced C3 to transplant injury was unknown. Current evidence indicates that defective local synthesis of C3 both reduces tissue injury and lowers the antidonor T cell response, substantially increasing graft survival. Among various possible explanations to account for these findings, the data favours a direct effect of complement on alloreactive T cell stimulation. Study of complement gene regulation by common immunosuppressive agents suggests that they do not influence local complement synthesis. Alternative approaches are therefore required to control the local effect of complement in the extravascular tissue compartment of the graft.


Subject(s)
Complement System Proteins/biosynthesis , Graft Rejection/immunology , Kidney Transplantation/immunology , Acute Disease , Animals , Complement C3c/biosynthesis , Complement C3c/immunology , Complement C4/biosynthesis , Complement C4/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Gene Expression Regulation , Graft Rejection/pathology , Humans , Lymphocyte Activation , Mice , Peptide Fragments/metabolism , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL