Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Clin Chem Lab Med ; 62(4): 674-681, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-37943101

ABSTRACT

OBJECTIVES: Tissue transglutaminase (tTG) IgA antibodies are a hallmark for celiac disease (CD). In CD patients on gluten free diet (GFD) these antibodies are transient. Few studies are available comparing the tTG-IgA assay characteristics for monitoring response to GFD. Since discrepant results were reported in patients on GFD after switching tTG-IgA assays, we conducted a retrospective observational study to monitor GFD response using three different tTG-IgA assays. METHODS: Diagnostic samples from 44 adults and 17 children with CD were included. Of most patients two follow-up samples after introduction of GFD were available. In all samples tTG-IgA were assessed using one fluorochrome-enzyme immuno-assay (FEIA) and two chemiluminescence immuno-assays (CLIA) and intestinal fatty acid binding protein (i-FABP) as surrogate marker for intestinal epithelial damage was measured. RESULTS: Using CLIA assays, normalization of antibody levels was delayed compared to FEIA (p<0.001). Of all samples taken after at least 6 months on GFD with elevated i-FABP indicating intestinal epithelial damage, 40 % had positive tTG-IgA according to the FEIA, 85 and 90 % according to the two CLIA. CONCLUSIONS: Normalization of tTG-IgA in patients on GFD depends on the assay used. Both CLIA appear to be more sensitive in detecting suboptimal treatment response in CD-indicated by elevated i-FABP - when applying the manufacturer's recommended cut-off for the diagnosis of CD.


Subject(s)
Celiac Disease , Child , Adult , Humans , Celiac Disease/diagnosis , Diet, Gluten-Free , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases , Autoantibodies , Immunoglobulin A
2.
Clin Chem Lab Med ; 61(8): 1446-1454, 2023 07 26.
Article in English | MEDLINE | ID: mdl-36857146

ABSTRACT

OBJECTIVES: Celiac disease (CD) is an immune-mediated enteropathy driven by gluten intake. Presence of tTG-IgA antibodies is important for the diagnosis. However, different tTG-IgA assays are used and test performance may vary. Therefore, a retrospective multicenter study was performed to compare the diagnostic performance of three assays. METHODS: The fluorescence enzyme-linked immunoassay (FEIA) EliA Celikey IgA (Phadia), the chemiluminescence immunoassays (CLIA) h-tTG IgA QUANTA Flash® (Inova Diagnostics) and the anti-tTG ChLIA IgA (Euroimmun) were compared. Diagnostic samples from CD cases (95 adults; 65 children) and controls (479 adults; 253 children) were included. Samples were blinded and reanalyzed on all platforms. RESULTS: A high quantitative correlation between platforms was found (p<0.0001). Both CLIA were more sensitive (adults 100%; children 100%) compared to the FEIA (adults 88.4%; children 96.6%). Specificity of all assays was high (≥97.6%) with the FEIA having the highest specificity. A cut-off based on receiver operator characteristic analysis (6.5 U/mL) improved the sensitivity of the FEIA (adults 95.8%; children 100%) without affecting specificity. Cut-off values for the CLIA assays did not need further optimization. With the FEIA, 71% of pediatric cases had a tTG-IgA level ≥10× upper limit of normal compared to 91 and 92% with QUANTA Flash and ChLIA, respectively. CONCLUSIONS: All platforms have high diagnostic accuracy. The CLIA assays are more sensitive compared to the FEIA assay. A lower cut-off for the FEIA improves diagnostic performance, particularly in adult cases that, as demonstrated in this study, present with lower tTG-IgA levels compared to pediatric cases.


Subject(s)
Celiac Disease , Transglutaminases , Adult , Humans , Child , Celiac Disease/diagnosis , Sensitivity and Specificity , Immunoassay , Immunoglobulin A , Autoantibodies
3.
Clin Chem Lab Med ; 60(7): 1046-1052, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35470638

ABSTRACT

OBJECTIVES: Diagnosis of type I hypersensitivity is based on anamnesis, provocation as well as blood- and skin testing. Multiplex specific IgE (sIgE) testing enables determination of sIgE antibodies against multiple recombinant or purified natural allergen components. The aim of this study was to evaluate the performance of the novel ALEX2® (Allergy Explorer, ALEX2 test introduced on the market November 2019) multiplex platform and to compare it with the ImmunoCAP ISAC® test system. METHODS: Serum samples of 49 patients, routinely determined with ISAC, were selected based on positive results covering in total most of the 112 ISAC components. Cohen's kappa, negative percent agreement (NPA), and positive percent agreement (PPA) of ALEX2 data compared to ISAC data (as a non-reference standard) were computed for those allergen components present on both platforms (n=103). Furthermore, in some samples sIgE results against allergen extracts and/or -components tested with either ImmunoCAP® (ThermoFisher) or IMMULITE® (Siemens) were available and compared to ALEX2 results. RESULTS: The overall agreement between ISAC and ALEX2 common allergen components was 94%. NPA and PPA were respectively 95 and 90%. Kappa values differed for specific allergen groups and varied between 0.60 and 0.92 showing moderate to almost perfect agreement. Of the qualitative discrepancies between ALEX2 and ISAC, 59% were related to weak positive results i.e. results under 1 kUA/L or 1 ISU, respectively. CONCLUSIONS: The method comparison between ISAC and ALEX2 multiplex tests showed a high concordance for those allergen components present on both platforms.


Subject(s)
Hypersensitivity , Immunoglobulin E , Allergens , Humans , Hypersensitivity/diagnosis , Skin Tests
4.
J Infect Dis ; 222(8): 1265-1269, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32726417

ABSTRACT

We determined and compared the humoral immune response in patients with severe (hospitalized) and mild (nonhospitalized) coronavirus disease 2019 (COVID-19). Patients with severe disease (n = 38) develop a robust antibody response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), including immunoglobulin G and immunoglobulin A antibodies. The geometric mean 50% virus neutralization titer is 1:240. SARS-CoV-2 infection was found in hospital personnel (n = 24), who developed mild symptoms necessitating leave of absence and self-isolation, but not hospitalization; 75% developed antibodies, but with low/absent virus neutralization (60% with titers <1:20). While severe COVID-19 patients develop a strong antibody response, mild SARS-CoV-2 infections induce a modest antibody response. Long-term monitoring will show whether these responses predict protection against future infections.


Subject(s)
Antibodies, Viral/immunology , Betacoronavirus/immunology , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Antibodies, Viral/blood , Antibody Formation , Betacoronavirus/isolation & purification , COVID-19 , Cohort Studies , Coronavirus Infections/blood , Coronavirus Infections/virology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Neutralization Tests , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/virology , SARS-CoV-2 , Severity of Illness Index
6.
Clin Chim Acta ; 476: 154-159, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29170107

ABSTRACT

BACKGROUND: Detection of antinuclear antibodies (ANA) by indirect immunofluorescence assay (IIFA) is increasingly substituted by fully automated solid phase immunoassays. This study evaluated the performance of an automated chemiluminescence immunoassay (CIA) and fluorescence enzyme immunoassay (FEIA) and compared their performance to that of IIFA. METHODS: The study included an unselected prospective study population suspected of systemic autoimmune rheumatic disease. ANA were measured by IIFA, while in parallel sera were tested by CIA QUANTA Flash CTD Screen Plus on the BIO-FLASH® and FEIA EliA CTD Screen on the Phadia® 250 system. As validation, retrospective cohorts of patients with ANA-associated rheumatic disease (AARD) and healthy controls were tested. RESULTS: Prospectively, sensitivity of IIFA, CIA and FEIA was 90%, 99% and 92%, respectively. Specificity was 76%, 76% and 84%, respectively. Total percent agreements between the three methods were 75.2% (IIFA vs. CIA), 79.2% (IIFA vs. FEIA) and 85.4% (FEIA vs. CIA). The AUC values were 0.95 for CIA and 0.93 for FEIA and did not significantly differ. Retrospectively in individual AARD cohorts, similar results were obtained comparing both CTD screens. CONCLUSIONS: Both FEIA and CIA CTD screen significantly outperformed IIFA, with a higher specificity for FEIA and higher sensitivity for CIA. Based on ROC analysis, major contributor to the difference between the two solid phase immunoassays was the cut-off.


Subject(s)
Antibodies, Antinuclear/analysis , Antigens, Nuclear/analysis , Arthritis, Rheumatoid/diagnosis , Automation , Fluoroimmunoassay , Luminescent Measurements , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Antigens, Nuclear/blood , Antigens, Nuclear/immunology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Humans , Prospective Studies
7.
Nephrol Dial Transplant ; 32(12): 2132-2141, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29045706

ABSTRACT

BACKGROUND: Kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) are promising biomarkers for monitoring delayed graft function (DGF) after kidney transplantation. Here we investigated localization and distribution of KIM-1 and NGAL staining in renal allograft biopsies and studied their association with histological features, functional DGF (fDGF) and the tubular function slope (TFS), a functioning proximal tubular epithelial cell (PTEC) marker. METHODS: Day 10 protocol biopsies of 64 donation after circulatory death recipients were stained for KIM-1 and NGAL and the positive area was quantified using ImageJ software. Biopsies were scored according to Banff and acute tubular necrosis (ATN) criteria. A 99mtechnetium-mercaptoacetyltriglycine (99mTc-MAG3)-renography was performed to calculate TFS. RESULTS: KIM-1 staining was located on the brush border of tubular epithelial cells (TECs) and correlated with denudation, while NGAL was present more focally in a cytoplasmic distribution. KIM-1 and NGAL staining were not correlated and no co-localization was observed. Quantitative stainings were not associated with fDGF, but KIM-1 tended to be higher in patients with prolonged fDGF (≥21 days; P = 0.062). No correlation was observed between the quantitative tissue stainings and urinary KIM-1 or NGAL. Quantitative KIM-1 staining was inversely correlated with the TFS (Spearman's ρ = -0.53; P < 0.001), whereas NGAL was not. The latter finding might be because cortical NGAL staining is dependent on filtration and subsequent reabsorption by functioning PTECs. Staining of NGAL was indeed restricted to PTECs, as shown by co-localization with a PTEC-specific lectin. CONCLUSIONS: KIM-1 and NGAL staining showed different localization and distribution. Quantitative KIM-1 staining was inversely correlated with functioning PTECs.


Subject(s)
Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Delayed Graft Function/diagnosis , Epithelial Cells/pathology , Hepatitis A Virus Cellular Receptor 1/metabolism , Kidney Transplantation/adverse effects , Kidney Tubules, Proximal/pathology , Aged , Animals , Biopsy , Delayed Graft Function/metabolism , Epithelial Cells/metabolism , Female , Humans , Kidney Tubules, Proximal/injuries , Kidney Tubules, Proximal/metabolism , Lipocalin-2/metabolism , Male , Middle Aged , Rats , Rats, Inbred Lew , Staining and Labeling , Transplantation, Homologous
8.
Am J Physiol Renal Physiol ; 310(11): F1308-16, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26984957

ABSTRACT

Previous studies have shown that complement activation on renal tubular cells is involved in the induction of interstitial fibrosis and cellular injury. Evidence suggests that the tubular cell damage is initiated by the alternative pathway (AP) of complement with properdin having an instrumental role. Properdin is a positive regulator of the AP, which can bind necrotic cells as well as viable proximal tubular epithelial cells (PTECs), inducing complement activation. Various studies have indicated that in the circulation there is an unidentified inhibitor of properdin. We investigated the ability of C-reactive protein (CRP), both in its monomeric (mCRP) and pentameric (pCRP) form, to inhibit AP activation and injury in vitro on renal tubular cells by fluorescent microscopy, ELISA, and flow cytometry. We demonstrated that preincubation of properdin with normal human serum inhibits properdin binding to viable PTECs. We identified mCRP as a factor able to bind to properdin in solution, thereby inhibiting its binding to PTECs. In contrast, pCRP exhibited no such binding and inhibitory effect. Furthermore, mCRP was able to inhibit properdin-directed C3 and C5b-9 deposition on viable PTECs. The inhibitory ability of mCRP was not unique for viable cells but also demonstrated for binding to necrotic Jurkat cells, a target for properdin binding and complement activation. In summary, mCRP is an inhibitor of properdin in both binding to necrotic cells and viable renal cells, regulating complement activation on the cell surface. We propose that mCRP limits amplification of tissue injury by controlling properdin-directed complement activation by damaged tissue and cells.


Subject(s)
C-Reactive Protein/metabolism , Complement Activation/drug effects , Complement C3/metabolism , Complement C5/metabolism , Kidney/metabolism , Properdin/pharmacology , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Kidney/drug effects , Protein Binding/drug effects
9.
Diabetologia ; 59(4): 853-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26768002

ABSTRACT

AIMS/HYPOTHESIS: High levels of circulating mannan-binding lectin (MBL) are associated with the development of diabetic nephropathy and hyperglycaemia-induced vasculopathy. Here, we aimed to assess the effect of glycaemic control on circulating levels of MBL and the relationship of these levels with vascular damage. METHODS: We assessed MBL levels and corresponding MBL2 genotype, together with vascular endothelial growth factor (VEGF) levels as a marker of vascular damage, in type 1 diabetes patients with diabetic nephropathy before and after simultaneous pancreas-kidney (SPK) transplantation. We included diabetic nephropathy patients (n = 21), SPK patients (n = 37), healthy controls (n = 19), type 1 diabetes patients (n = 15) and diabetic nephropathy patients receiving only a kidney transplant (n = 15). Fourteen diabetic nephropathy patients were followed up for 12 months after SPK. RESULTS: We found elevated circulating MBL levels in diabetic nephropathy patients, and a trend towards elevated circulating MBL levels in type 1 diabetes patients, compared with healthy control individuals. MBL levels in SPK patients completely normalised and our data indicate that this predominantly occurs in patients with a polymorphism in the MBL2 gene. By contrast, MBL levels in kidney transplant only patients remained elevated, suggesting that glycaemic control but not reversal of renal failure is associated with decreased MBL levels. In line, levels of glucose and HbA1c, but not creatinine levels and estimated GFR, were correlated with MBL levels. VEGF levels were associated with levels of MBL and HbA1c in an MBL-polymorphism-dependent manner. CONCLUSIONS/INTERPRETATION: Taken together, circulating MBL levels are associated with diabetic nephropathy and are dependent on glycaemic control, possibly in an MBL2-genotype-dependent manner.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/surgery , Kidney Transplantation , Mannose-Binding Lectin/blood , Pancreas Transplantation , Vascular Endothelial Growth Factor A/blood , Cross-Sectional Studies , Diabetes Mellitus, Type 1/blood , Female , Genotype , Humans , Male , Mannose-Binding Lectin/genetics , Vascular Endothelial Growth Factor A/genetics
10.
PLoS One ; 10(9): e0137795, 2015.
Article in English | MEDLINE | ID: mdl-26367533

ABSTRACT

Preoperative fasting and dietary restriction offer robust protection against renal ischemia/reperfusion injury (I/RI) in mice. We recently showed that Mannan-binding lectin (MBL), the initiator of the lectin pathway of complement activation, plays a pivotal role in renal I/RI. Based on these findings, we investigated the effect of short-term DR (30% reduction of total food intake) or three days of water only fasting on MBL in 10-12 weeks old male C57/Bl6 mice. Both dietary regimens significantly reduce the circulating levels of MBL as well as its mRNA expression in liver, the sole production site of MBL. Reconstitution of MBL abolished the protection afforded by dietary restriction, whereas in the fasting group the protection persisted. These data show that modulation of MBL is involved in the protection against renal I/RI induced by dietary restriction, and suggest that the mechanisms of protection induced by dietary restriction and fasting may be different.


Subject(s)
Caloric Restriction , Gene Expression Regulation , Kidney Diseases/prevention & control , Liver/metabolism , Mannose-Binding Lectin/biosynthesis , Reperfusion Injury/prevention & control , Animals , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Liver/pathology , Male , Mice , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
11.
J Biol Chem ; 289(51): 35421-30, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25349208

ABSTRACT

Lipoproteins can induce complement activation resulting in opsonization and binding of these complexes to complement receptors. We investigated the binding of opsonized native LDL and acetylated LDL (acLDL) to the complement receptor 1 (CR1). Binding of complement factors C3b, IgM, C1q, mannose-binding lectin (MBL), and properdin to LDL and acLDL were investigated by ELISA. Subsequent binding of opsonized LDL and acLDL to CR1 on CR1-transfected Chinese Hamster Ovarian cells (CHO-CR1) was tested by flow cytometry. Both native LDL and acLDL induced complement activation with subsequent C3b opsonization upon incubation with normal human serum. Opsonized LDL and acLDL bound to CR1. Binding to CHO-CR1 was reduced by EDTA, whereas MgEGTA only reduced the binding of opsonized LDL, but not of acLDL suggesting involvement of the alternative pathway in the binding of acLDL to CR1. In vitro incubations showed that LDL bound C1q, whereas acLDL bound to C1q, IgM, and properdin. MBL did neither bind to LDL nor to acLDL. The relevance of these findings was demonstrated by the fact that ex vivo up-regulation of CR1 on leukocytes was accompanied by a concomitant increased binding of apolipoprotein B-containing lipoproteins to leukocytes without changes in LDL-receptor expression. In conclusion, CR1 is able to bind opsonized native LDL and acLDL. Binding of LDL to CR1 is mediated via the classical pathway, whereas binding of acLDL is mediated via both the classical and alternative pathways. Binding of lipoproteins to CR1 may be of clinical relevance due to the ubiquitous cellular distribution of CR1.


Subject(s)
Complement Activation , Complement C3b/metabolism , Lipoproteins, LDL/metabolism , Receptors, Complement 3b/metabolism , Animals , Apolipoproteins B/metabolism , CHO Cells , Cells, Cultured , Complement C1q/metabolism , Complement Pathway, Alternative , Complement Pathway, Classical , Cricetinae , Cricetulus , Edetic Acid/pharmacology , Flow Cytometry , Humans , Immunoglobulin M/metabolism , Leukocytes/cytology , Leukocytes/drug effects , Leukocytes/metabolism , Lipopolysaccharides/pharmacology , Opsonin Proteins/metabolism , Properdin/metabolism , Protein Binding/drug effects , Receptors, Complement 3b/genetics
12.
J Am Soc Nephrol ; 25(8): 1710-22, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24610930

ABSTRACT

Ischemia/reperfusion injury (IRI) is a central phenomenon in kidney transplantation and AKI. Integrity of the renal peritubular capillary network is an important limiting factor in the recovery from IRI. MicroRNA-126 (miR-126) facilitates vascular regeneration by functioning as an angiomiR and by modulating mobilization of hematopoietic stem/progenitor cells. We hypothesized that overexpression of miR-126 in the hematopoietic compartment could protect the kidney against IRI via preservation of microvascular integrity. Here, we demonstrate that hematopoietic overexpression of miR-126 increases neovascularization of subcutaneously implanted Matrigel plugs in mice. After renal IRI, mice overexpressing miR-126 displayed a marked decrease in urea levels, weight loss, fibrotic markers, and injury markers (such as kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin). This protective effect was associated with a higher density of the peritubular capillary network in the corticomedullary junction and increased numbers of bone marrow-derived endothelial cells. Hematopoietic overexpression of miR-126 increased the number of circulating Lin(-)/Sca-1(+)/cKit(+) hematopoietic stem and progenitor cells. Additionally, miR-126 overexpression attenuated expression of the chemokine receptor CXCR4 on Lin(-)/Sca-1(+)/cKit(+) cells in the bone marrow and increased renal expression of its ligand stromal cell-derived factor 1, thus favoring mobilization of Lin(-)/Sca-1(+)/cKit(+) cells toward the kidney. Taken together, these results suggest overexpression of miR-126 in the hematopoietic compartment is associated with stromal cell-derived factor 1/CXCR4-dependent vasculogenic progenitor cell mobilization and promotes vascular integrity and supports recovery of the kidney after IRI.


Subject(s)
Acute Kidney Injury/prevention & control , Hematopoietic Stem Cells/physiology , Kidney/blood supply , MicroRNAs/physiology , Neovascularization, Physiologic/physiology , Reperfusion Injury/prevention & control , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Cell Movement/physiology , Chemokine CXCL12/metabolism , Kidney/metabolism , Kidney/pathology , Male , Mice, Inbred C57BL , Receptors, CXCR4/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
13.
Am J Physiol Renal Physiol ; 305(6): F901-10, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23825073

ABSTRACT

Endothelial cells (ECs) are highly susceptible to hypoxia and easily affected upon ischemia-reperfusion (I/R) during renal transplantation. Pericytes and angiopoeitins play important role in modulating EC function. In the present study, we investigate the effect of renal I/R on the dynamics of angiopoietin expression and its association with pericytes and fibrosis development. Male Lewis rats were subjected to unilateral renal ischemia for 45 min followed by removal of the contralateral kidney. Rats were killed at different time points after reperfusion. Endothelial integrity (RECA-1), pericytes [platelet-derived growth factor receptor-ß (PDGFR-ß)], angiopoietin-2 (Ang-2)/angiopoietin-1 (Ang-1) expression, and interstitial collagen deposition (Sirius red and α-smooth muscle actin) were assessed using immunohistochemistry and RT-PCR. Our study shows an increase in protein expression of Ang-2 starting at 5 h and remaining elevated up to 72 h, with a consequently higher Ang-2/Ang-1 ratio after renal I/R (P < 0.05 at 48 h). This was accompanied by an increase in protein expression of the pericytic marker PDGFR-ß and a loss of ECs (both at 72 h after I/R, P < 0.05). Nine weeks after I/R, when renal function was restored, we observed normalization of the Ang-2/Ang-1 ratio and PDGFR-ß expression and increase in cortical ECs, which was accompanied by fibrosis. Renal I/R induces a dysbalance of Ang-2/Ang-1 accompanied by proliferation of pericytes, EC loss, and development of fibrosis. The Ang-2/Ang-1 balance was reversed to baseline at 9 wk after renal I/R, which coincided with restoration of cortical ECs and pericytes. Our findings suggest that angiopoietins and pericytes play an important role in renal microvascular remodeling and development of fibrosis.


Subject(s)
Angiopoietins/metabolism , Pericytes/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Angiopoietin-1/biosynthesis , Angiopoietin-2/biosynthesis , Animals , Endothelial Cells , Fibrosis , Kidney Diseases/pathology , Male , Pericytes/metabolism , Rats , Rats, Inbred Lew , Receptor, Platelet-Derived Growth Factor beta/biosynthesis
14.
Transplantation ; 95(6): 816-20, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23348894

ABSTRACT

BACKGROUND: Ischemia/reperfusion (I/R) injury has a major impact on kidney graft function and survival. Animal studies have suggested a role for complement activation in mediating I/R injury; however, results are not unambiguous. Whether complement activation is involved in clinical I/R injury in humans is still unclear. METHODS: In the present study, we assessed the formation and release of C5b-9 during early reperfusion in clinical kidney transplantation in living donor, brain-dead donor, and cardiac dead donor kidney transplantation. By arteriovenous measurements and histologic studies, local terminal complement activation in the reperfused kidney was assessed. RESULTS: There was no release of soluble C5b-9 (sC5b-9) from living donor kidneys, nor was there a release of C5a. In contrast, instantly after reperfusion, there was a significant but transient venous release of sC5b-9 from the reperfused kidney graft in brain-dead donor and cardiac dead donor kidney transplantation. This short-term activation of the terminal complement cascade in deceased-donor kidney transplantation was not reflected by renal tissue deposition of C5b-9 in biopsies taken 45 min after reperfusion. CONCLUSIONS: This systematic study in human kidney transplantation shows an acute but nonsustained sC5b-9 release on reperfusion in deceased-donor kidney transplantation. This instantaneous, intravascular terminal complement activation may be induced by intravascular cellular debris and hypoxic or injured endothelium.


Subject(s)
Complement Membrane Attack Complex/metabolism , Kidney Transplantation/methods , Reperfusion/methods , Acute Disease , Adult , Aged , Animals , Biopsy/methods , Complement Activation , Female , Humans , Immunohistochemistry/methods , Kidney/anatomy & histology , Male , Middle Aged , Models, Statistical , Reperfusion Injury/therapy , Tissue Donors
15.
Mol Immunol ; 53(1-2): 132-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22906481

ABSTRACT

Previous experiments from our laboratories have identified peptides derived from the human astrovirus coat protein (CP) that bind C1q and mannose binding lectin (MBL) inhibiting activation of the classical and lectin pathways of complement, respectively. The purpose of this study was to evaluate the function of these coat protein peptides (CPPs) in an in vitro model of complement-mediated disease (ABO incompatibility), preliminarily assess their in vivo complement suppression profile and develop more highly potent derivatives of these molecules. E23A, a 30 amino acid CPP derivative previously demonstrated to inhibit classical pathway activation was able to dose-dependently inhibit lysis of AB erythrocytes treated with mismatched human O serum. Additionally, when injected into rats, E23A inhibited the animals' serum from lysing antibody-sensitized erythrocytes, providing preliminary in vivo functional evidence that this CPP can cross the species barrier to inhibit serum complement activity in rodents. A rational drug design approach was implemented to identify more potent CPP derivatives, resulting in the identification and characterization of a 15 residue peptide (polar assortant (PA)), which demonstrated both superior inhibition of classical complement pathway activation and robust binding to C1q collagen-like tails. PA also inhibited ABO incompatibility in vitro and demonstrated in vivo complement suppression up to 24h post-injection. CPP's ability to inhibit ABO incompatibility in vitro, proof of concept in vivo inhibitory activity in rats and the development of the highly potent PA derivative set the stage for preclinical testing of this molecule in small animal models of complement-mediated disease.


Subject(s)
Blood Group Incompatibility/drug therapy , Capsid Proteins/pharmacology , Complement Pathway, Classical/drug effects , Complement Pathway, Mannose-Binding Lectin/drug effects , Peptides/pharmacology , ABO Blood-Group System/immunology , Amino Acid Sequence , Animals , Capsid Proteins/chemistry , Humans , Peptides/chemistry , Rats , Surface Plasmon Resonance
16.
Transplantation ; 94(5): 478-85, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22892990

ABSTRACT

BACKGROUND: Innate immunity plays a role in controlling adaptive immune responses. METHODS: We investigated the clinical relevance of single nucleotide polymorphisms in 22 genes encoding innate, secreted, and signaling pattern recognition receptors in a total of 520 donor-recipient pairs of postmortem, human leukocyte antigen-DR-compatible kidney transplantations. Associations with rejection incidence were tested in an a priori randomized training set and validation set. RESULTS: Polymorphisms in TLR-3 (rs3775296) in the recipients and in ficolin-2 (rs7851696; Ala258Ser) and C1qR1 (rs7492) in the donors showed the strongest association with severe rejection. In multivariate analysis, presence of the ficolin-2 Ala258Ser variant in the donor predicted lower incidence of severe rejection (odds ratio=0.3; 95% confidence interval, 0.1-0.9; P=0.024) and of graft loss (hazard ratio=0.5; 95% confidence interval, 0.2-1.0; P=0.046) independently of clinical risk factors. Ficolin-2 messenger RNA expression was detected in pretransplantation biopsies from 69 donor grafts. Serum and tissue ficolin-2 levels were unaffected by genotype. Ficolin-2 protein, which bound to dying cells, was detected in donor kidneys in a passenger leukocyte-like pattern. Indeed, monocytes, monocyte-derived macrophages, and peripheral blood mononuclear cells expressed ficolin-2. Donor grafts with the ficolin-2 Ala258Ser variant contained significantly elevated expression of interleukin 6, having ascribed cytoprotective effects. It has been described that Ala258Ser leads to increased binding capacity of ficolin-2 to N-acetylglucosamine. CONCLUSIONS: Presence of the ficolin-2 Ala258Ser polymorphism in the donor independently predicts improved graft outcome. Based on mechanistic data, we propose that this functional polymorphism leads to more efficient handling of injured cells by phagocytozing cells, resulting in decreased intragraft exposure to danger signals and dampened alloimmune responses.


Subject(s)
Graft Rejection/genetics , Graft Survival , Immunity, Innate/genetics , Kidney Transplantation , Lectins/genetics , Polymorphism, Single Nucleotide , Tissue Donors , Apoptosis , Biopsy , Exons , Gene Expression Regulation , Genotype , Graft Rejection/blood , Graft Rejection/immunology , Graft Rejection/metabolism , Humans , Interleukin-1beta/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Jurkat Cells , Kaplan-Meier Estimate , Kidney Transplantation/adverse effects , Kidney Transplantation/immunology , Lectins/blood , Logistic Models , Multivariate Analysis , Netherlands , Odds Ratio , Phenotype , Proportional Hazards Models , Risk Assessment , Risk Factors , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Time Factors , Treatment Outcome , Tumor Necrosis Factor-alpha/genetics , Ficolins
17.
J Biol Chem ; 287(37): 31471-81, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22815489

ABSTRACT

During proteinuria, renal tubular epithelial cells become exposed to ultrafiltrate-derived serum proteins, including complement factors. Recently, we showed that properdin binds to tubular heparan sulfates (HS). We now document that factor H also binds to tubular HS, although to a different epitope than properdin. Factor H was present on the urinary side of renal tubular cells in proteinuric, but not in normal renal tissues and colocalized with properdin in proteinuric kidneys. Factor H dose-dependently bound to proximal tubular epithelial cells (PTEC) in vitro. Preincubation of factor H with exogenous heparin and pretreatment of PTECs with heparitinase abolished the binding to PTECs. Surface plasmon resonance experiments showed high affinity of factor H for heparin and HS (K(D) values of 32 and 93 nm, respectively). Using a library of HS-like polysaccharides, we showed that chain length and high sulfation density are the most important determinants for glycosaminoglycan-factor H interaction and clearly differ from properdin-heparinoid interaction. Coincubation of properdin and factor H did not hamper HS/heparin binding of one another, indicating recognition of different nonoverlapping epitopes on HS/heparin by factor H and properdin. Finally we showed that certain low anticoagulant heparinoids can inhibit properdin binding to tubular HS, with a minor effect on factor H binding to tubular HS. As a result, these heparinoids can control the alternative complement pathway. In conclusion, factor H and properdin interact with different HS epitopes of PTECs. These interactions can be manipulated with some low anticoagulant heparinoids, which can be important for preventing complement-derived tubular injury in proteinuric renal diseases.


Subject(s)
Complement Factor H/metabolism , Epitopes/metabolism , Heparitin Sulfate/metabolism , Kidney Tubules/metabolism , Properdin/metabolism , Proteinuria/metabolism , Animals , Cell Line , Humans , Kidney Tubules/pathology , Male , Protein Binding , Proteinuria/mortality , Rats , Rats, Wistar
18.
Transpl Immunol ; 27(1): 55-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22709941

ABSTRACT

Local activation of the complement system has been associated with ischemia/reperfusion injury following kidney transplantation and tubular injury under proteinuric conditions. The soluble terminal complement complex sC5b-9 is a stable end-product of the complement cascade, and as such a promising urinary biomarker. In the early post-transplant period we found high urinary levels of sC5b-9, significantly correlating with the degree of proteinuria, suggesting activation of filtered complement components at the tubular epithelial surface of the kidney. However, when mimicking proteinuria in vitro by exposing serum (or blood) to urine (both negative for sC5b-9), we found extensive generation of sC5b-9 in urine. This process was inhibited by EDTA, confirming activation of the complement system. In conclusion, although sC5b-9 is an attractive urinary biomarker, one should be aware of the risk of extra-renal complement activation independent of a renal contribution. This may be of special interest when measuring urinary sC5b-9 following kidney transplantation in which procedure-related (microscopic) hematuria and proteinuria are common.


Subject(s)
Complement Membrane Attack Complex/urine , Kidney Transplantation , Reperfusion Injury/urine , Biomarkers/urine , Edetic Acid/pharmacology , Female , Hematuria , Humans , Male , Middle Aged , Proteinuria
19.
Eur J Cardiothorac Surg ; 41(5): e117-25, 2012 May.
Article in English | MEDLINE | ID: mdl-22408044

ABSTRACT

OBJECTIVES: Complement activation is considered an important mediator of myocardial ischaemia/reperfusion (I/R) injury. Although complement inhibitors are highly effective in animals, clinical trials fail to show a substantial benefit in humans. This raises questions on the role of complement activation in human myocardial I/R injury. METHODS: Soluble C5b-9, i.e. terminal complement complex, and C5a were assessed in patients with non-ischaemic (n = 10) and ischaemic heart failure (n = 10), and patients without heart failure (n = 10) undergoing cardiac surgery. To study the pathophysiology of human I/R injury, a model of arteriovenous measurements over the reperfused heart was applied at consecutive time points during the early reperfusion phase. Furthermore, C3d and C5b-9 depositions in pre-reperfusion myocardial and endomyocardial tissue were evaluated and compared to pre-transplantation tissue from myocardial allografts. RESULTS: Simultaneous assessment of soluble C5b-9 and C5a in systemical and myocardial venous blood samples revealed the absence of net release from the reperfused heart in all three patient groups. Biopsies of patients with non-ischaemic heart failure showed the most abundant myocardial depositions of C3d and C5b-9: 4.8 times more C3d (P = 0.008) and 4.7 times more C5b-9 (P = 0.004) than donor tissue. Also C3d was abundantly present in endomyocardial tissue of both heart failure groups compared to donors (both P = 0.02). CONCLUSIONS: No evidence was obtained that terminal complement activation is involved in the acute phase following myocardial reperfusion. Since complement deposition was already present before reperfusion, human complement inhibition might be more beneficial in the preoperative phase than during reperfusion.


Subject(s)
Cardiac Surgical Procedures/adverse effects , Complement Activation/physiology , Myocardial Reperfusion Injury/immunology , Aged , Cardiopulmonary Bypass/adverse effects , Complement C3d/metabolism , Complement C5/metabolism , Heart Failure/immunology , Humans , Middle Aged , Myocardial Reperfusion , Myocardial Reperfusion Injury/etiology , Myocardium/immunology
20.
Mol Immunol ; 49(1-2): 75-83, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21855148

ABSTRACT

Various studies indicate a role for both anti-nucleosome and anti-C1q autoantibodies in glomerulonephritis in patients with systemic lupus erythematosus. However, a causal relationship between these autoantibodies and the development of lupus nephritis has not been fully established. Since injury of the endothelium is a major target in lupus nephritis we assessed the interaction of C1q and nucleosomes with glomerular endothelial cells in vitro in the presence or absence of autoantibodies against these antigens. We demonstrate a direct and dose-dependent binding of both nucleosomes and C1q to immortalized human glomerular endothelial cells (GEnC) in vitro, which in part is mediated by cell surface heparan sulfate. We demonstrate that nucleosomes and C1q serve as targets for monoclonal and polyclonal antibodies as well as for anti-nuclear autoantibodies from patients with systemic lupus erythematosus. An additive effect of anti-C1q autoantibodies on anti-nucleosome mediated complement activation was observed. Furthermore, we showed that the activation of complement on glomerular endothelial cells is mediated by the classical pathway since the deposition of C3 on GEnC is abrogated by MgEGTA and does not occur in C1q-depleted serum. Taken together, our studies demonstrate a direct binding of both nucleosomes and C1q to glomerular endothelial cells in vitro. The subsequent binding of autoantibodies against nucleosomes in patients with systemic lupus erythematosus is potentially pathogenic and autoantibodies against C1q seem to have an additional effect.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Complement C1q/immunology , Kidney Glomerulus/immunology , Lupus Nephritis/immunology , Nucleosomes/immunology , Cell Separation , Complement Activation/immunology , Endothelial Cells/immunology , Flow Cytometry , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...