Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Transl Med ; 22(1): 17, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38178089

ABSTRACT

BACKGROUND: Hemolysis is a cardinal feature of hemolytic uremic syndrome (HUS) and during hemolysis excess arginase 1 is released from red blood cells. Increased arginase activity leads to reduced L-arginine, as it is converted to urea and L-ornithine, and thereby reduced nitric oxide bioavailability, with secondary vascular injury. The objective of this study was to investigate arginase release in HUS patients and laboratory models and correlate arginase levels to hemolysis and kidney injury. METHODS: Two separate cohorts of patients (n = 47 in total) with HUS associated with Shiga toxin-producing enterohemorrhagic E. coli (EHEC) and pediatric controls (n = 35) were investigated. Two mouse models were used, in which mice were either challenged intragastrically with E. coli O157:H7 or injected intraperitoneally with Shiga toxin 2. An in vitro model of thrombotic microangiopathy was developed in which Shiga toxin 2- and E. coli O157 lipopolysaccharide-stimulated human blood cells combined with ADAMTS13-deficient plasma were perfused over glomerular endothelial cells. Two group statistical comparisons were performed using the Mann-Whitney test, multiple groups were compared using the Kruskal-Wallis test followed by Dunn's procedure, the Wilcoxon signed rank test was used for paired data, or linear regression for continuous variables. RESULTS: HUS patients had excessively high plasma arginase 1 levels and activity (conversion of L-arginine to urea and L-ornithine) during the acute phase, compared to remission and controls. Arginase 1 levels correlated with lactate dehydrogenase activity, indicating hemolysis, as well as the need for dialysis treatment. Patients also exhibited high levels of plasma alpha-1-microglobulin, a heme scavenger. Both mouse models exhibited significantly elevated plasma arginase 1 levels and activity. Plasma arginase 1 levels correlated with lactate dehydrogenase activity, alpha-1-microglobulin and urea levels, the latter indicative of kidney dysfunction. In the in vitro model of thrombotic microangiopathy, bioactive arginase 1 was released and levels correlated to the degree of hemolysis. CONCLUSIONS: Elevated red blood cell-derived arginase was demonstrated in HUS patients and in relevant in vivo and in vitro models. The excessively high arginase levels correlated to the degree of hemolysis and kidney dysfunction. Thus, arginase inhibition should be investigated in HUS.


Subject(s)
Escherichia coli Infections , Escherichia coli O157 , Hemolytic-Uremic Syndrome , Renal Insufficiency , Thrombotic Microangiopathies , Humans , Child , Animals , Mice , Shiga Toxin 2 , Endothelial Cells , Hemolysis , Arginase , Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/therapy , Erythrocytes , Thrombotic Microangiopathies/complications , Urea , Arginine , Ornithine , Lactate Dehydrogenases , Escherichia coli Infections/complications , Escherichia coli Infections/therapy
2.
Pediatr Nephrol ; 39(4): 1105-1111, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37955705

ABSTRACT

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) can be associated with mutations, deletions, or hybrid genes in factor H-related (FHR) proteins. METHODS: A child with aHUS was investigated. Genetics was assessed by Sanger and next generation sequencing. Serum FHR5 was evaluated by immunoblotting, ELISA, and by induction of rabbit red blood cell hemolysis in the presence/absence of recombinant human rFHR5. Mutagenesis was performed in HEK cells. RESULTS: A heterozygous genetic variant in factor H-related protein 5 (CFHR5), M514R, was found in the child, who also had a homozygous deletion of CFHR3/CFHR1, and antibodies to factor H, as well as low levels of C3. Patient serum exhibited low levels of FHR5. In the presence of rabbit red blood cells, patient serum induced hemolysis which decreased when rFHR5 was added at physiological concentrations. Similar results were obtained using serum from the father, bearing the CFHR5 variant without factor H antibodies. Patient FHR5 formed normal dimers. The CFHR5 M514R variant was expressed in HEK cells and minimal secretion was detected whereas the protein level was elevated in cell lysates. CONCLUSIONS: Decreased secretion of the product of the mutant allele could explain the low FHR5 levels in patient serum. Reduced hemolysis when rFHR5 was added to serum suggests a regulatory role regarding complement activation on red blood cells. As such, low levels of FHR5, as demonstrated in the patient, may contribute to complement activation.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Child , Animals , Humans , Rabbits , Atypical Hemolytic Uremic Syndrome/genetics , Complement Factor H/genetics , Hemolysis , Homozygote , Sequence Deletion , Complement System Proteins , Antibodies
3.
Front Immunol ; 14: 1254759, 2023.
Article in English | MEDLINE | ID: mdl-37744338

ABSTRACT

Background: Complement activation in atypical hemolytic uremic syndrome (aHUS), C3 glomerulonephropathy (C3G) and immune complex-mediated membranoproliferative glomerulonephritis (IC-MPGN) may be associated with rare genetic variants. Here we describe gene variants in the Swedish and Norwegian populations. Methods: Patients with these diagnoses (N=141) were referred for genetic screening. Sanger or next-generation sequencing were performed to identify genetic variants in 16 genes associated with these conditions. Nonsynonymous genetic variants are described when they have a minor allele frequency of <1% or were previously reported as being disease-associated. Results: In patients with aHUS (n=94, one also had IC-MPGN) 68 different genetic variants or deletions were identified in 60 patients, of which 18 were novel. Thirty-two patients had more than one genetic variant. In patients with C3G (n=40) 29 genetic variants, deletions or duplications were identified in 15 patients, of which 9 were novel. Eight patients had more than one variant. In patients with IC-MPGN (n=7) five genetic variants were identified in five patients. Factor H variants were the most frequent in aHUS and C3 variants in C3G. Seventeen variants occurred in more than one condition. Conclusion: Genetic screening of patients with aHUS, C3G and IC-MPGN is of paramount importance for diagnostics and treatment. In this study, we describe genetic assessment of Nordic patients in which 26 novel variants were found.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Kidney Diseases , Humans , Complement System Proteins/genetics , Complement Activation/genetics , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/genetics , Gene Frequency
5.
Gut Microbes ; 14(1): 2122667, 2022.
Article in English | MEDLINE | ID: mdl-36138514

ABSTRACT

Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC) cause gastrointestinal infection and, in severe cases, hemolytic uremic syndrome which may lead to death. There is, to-date, no therapy for this infection. Stx induces ATP release from host cells and ATP signaling mediates its cytotoxic effects. Apyrase cleaves and neutralizes ATP and its effect on Stx and EHEC infection was therefore investigated. Apyrase decreased bacterial RecA and dose-dependently decreased toxin release from E. coli O157:H7 in vitro, demonstrated by reduced phage DNA and protein levels. The effect was investigated in a mouse model of E. coli O157:H7 infection. BALB/c mice infected with Stx2-producing E. coli O157:H7 were treated with apyrase intraperitoneally, on days 0 and 2 post-infection, and monitored for 11 days. Apyrase-treated mice developed disease two days later than untreated mice. Untreated infected mice lost significantly more weight than those treated with apyrase. Apyrase-treated mice exhibited less colonic goblet cell depletion and apoptotic cells, as well as lower fecal ATP and Stx2, compared to untreated mice. Apyrase also decreased platelet aggregation induced by co-incubation of human platelet-rich-plasma with Stx2 and E. coli O157 lipopolysaccharide in the presence of collagen. Thus, apyrase had multiple protective effects, reducing RecA levels, stx2 and toxin release from EHEC, reducing fecal Stx2 and protecting mouse intestinal cells, as well as decreasing platelet activation, and could thereby delay the development of disease.


Subject(s)
Bacteriophages , Escherichia coli Infections , Escherichia coli O157 , Gastrointestinal Microbiome , Adenosine Triphosphate/metabolism , Animals , Apyrase/metabolism , Apyrase/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli O157/genetics , Humans , Lipopolysaccharides/metabolism , Mice , Mice, Inbred BALB C , Shiga Toxin/metabolism , Shiga Toxin/pharmacology , Shiga Toxin 2/genetics , Shiga Toxin 2/metabolism , Shiga Toxin 2/pharmacology
6.
Front Immunol ; 13: 807959, 2022.
Article in English | MEDLINE | ID: mdl-35250980

ABSTRACT

Shiga toxin-producing Escherichia coli O157:H7 is a virulent strain causing severe gastrointestinal infection, hemolytic uremic syndrome and death. To date there are no specific therapies to reduce progression of disease. Here we investigated the effect of pooled immunoglobulins (IgG) on the course of disease in a mouse model of intragastric E. coli O157:H7 inoculation. Intraperitoneal administration of murine IgG on day 3, or both on day 3 and 6, post-inoculation improved survival and decreased intestinal and renal pathology. When given on both day 3 and 6 post-inoculation IgG treatment also improved kidney function in infected mice. Murine and human commercially available IgG preparations bound to proteins in culture filtrates from E. coli O157:H7. Bound proteins were extracted from membranes and peptide sequences were identified by mass spectrometry. The findings showed that murine and human IgG bound to E. coli extracellular serine protease P (EspP) in the culture filtrate, via the IgG Fc domain. These results were confirmed using purified recombinant EspP and comparing culture filtrates from the wild-type E. coli O157:H7 strain to a deletion mutant lacking espP. Culture filtrates from wild-type E. coli O157:H7 exhibited enzymatic activity, specifically associated with the presence of EspP and demonstrated as pepsin cleavage, which was reduced in the presence of murine and human IgG. EspP is a virulence factor previously shown to promote colonic cell injury and the uptake of Shiga toxin by intestinal cells. The results presented here suggest that IgG binds to EspP, blocks its enzymatic activity, and protects the host from E. coli O157:H7 infection, even when given post-inoculation.


Subject(s)
Escherichia coli Infections , Escherichia coli O157 , Escherichia coli Proteins , Immunoglobulin G , Serine Proteases , Animals , Escherichia coli Proteins/metabolism , Immunoglobulin G/metabolism , Mice , Serine/metabolism , Serine Endopeptidases/metabolism , Serine Proteases/metabolism
7.
Front Immunol ; 12: 690821, 2021.
Article in English | MEDLINE | ID: mdl-34177949

ABSTRACT

Complement factor B (FB) mutant variants are associated with excessive complement activation in kidney diseases such as atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathy and membranoproliferative glomerulonephritis (MPGN). Patients with aHUS are currently treated with eculizumab while there is no specific treatment for other complement-mediated renal diseases. In this study the phenotype of three FB missense variants, detected in patients with aHUS (D371G and E601K) and MPGN (I242L), was investigated. Patient sera with the D371G and I242L mutations induced hemolysis of sheep erythrocytes. Mutagenesis was performed to study the effect of factor D (FD) inhibition on C3 convertase-induced FB cleavage, complement-mediated hemolysis, and the release of soluble C5b-9 from glomerular endothelial cells. The FD inhibitor danicopan abrogated C3 convertase-associated FB cleavage to the Bb fragment in patient serum, and of the FB constructs, D371G, E601K, I242L, the gain-of-function mutation D279G, and the wild-type construct, in FB-depleted serum. Furthermore, the FD-inhibitor blocked hemolysis induced by the D371G and D279G gain-of-function mutants. In FB-depleted serum the D371G and D279G mutants induced release of C5b-9 from glomerular endothelial cells that was reduced by the FD-inhibitor. These results suggest that FD inhibition can effectively block complement overactivation induced by FB gain-of-function mutations.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Complement Activation , Complement Factor B/genetics , Complement Factor D/antagonists & inhibitors , Glomerulonephritis, Membranoproliferative/immunology , Animals , Atypical Hemolytic Uremic Syndrome/genetics , Child , Complement C3-C5 Convertases/immunology , Complement C3b/immunology , Complement Factor B/immunology , Complement Factor D/immunology , Endothelial Cells/immunology , Erythrocytes , Female , Glomerulonephritis, Membranoproliferative/genetics , Hemolysis , Humans , Infant , Kidney Glomerulus/cytology , Male , Middle Aged , Mutation , Phenotype , Rabbits , Sheep
8.
Article in English | MEDLINE | ID: mdl-32523894

ABSTRACT

Shiga toxin is the main virulence factor of non-invasive enterohemorrhagic Escherichia coli strains capable of causing hemolytic uremic syndrome. Our group has previously shown that the toxin can reach the kidney within microvesicles where it is taken up by renal cells and the vesicles release their cargo intracellularly, leading to toxin-mediated inhibition of protein synthesis and cell death. The aim of this study was to examine if recipient cells must express the globotriaosylceramide (Gb3) toxin receptor for this to occur, or if Gb3-negative cells are also susceptible after uptake of Gb3-positive and toxin-positive microvesicles. To this end we generated Gb3-positive A4GALT-transfected CHO cells, and a vector control lacking Gb3 (CHO-control cells), and decreased Gb3 synthesis in native HeLa cells by exposing them to the glycosylceramide synthase inhibitor PPMP. We used these cells, and human intestinal DLD-1 cells lacking Gb3, and exposed them to Shiga toxin 2-bearing Gb3-positive microvesicles derived from human blood cells. Results showed that only recipient cells that possessed endogenous Gb3 (CHO-Gb3 transfected and native HeLa cells) exhibited cellular injury, reduced cell metabolism and protein synthesis, after uptake of toxin-positive microvesicles. In Gb3-positive cells the toxin introduced via vesicles followed the retrograde pathway and was inhibited by the retrograde transport blocker Retro-2.1. CHO-control cells, HeLa cells treated with PPMP and DLD-1 cells remained unaffected by toxin-positive microvesicles. We conclude that Shiga toxin-containing microvesicles can be taken up by Gb3-negative cells but the recipient cell must express endogenous Gb3 for the cell to be susceptible to the toxin.


Subject(s)
Hemolytic-Uremic Syndrome , Shiga Toxin , Animals , Cricetinae , Cricetulus , HeLa Cells , Humans , Shiga Toxin 2
9.
Sci Rep ; 9(1): 14362, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31591425

ABSTRACT

Shiga toxin (Stx) is the main virulence factor of enterohemorrhagic Escherichia coli (EHEC), that cause gastrointestinal infection leading to hemolytic uremic syndrome. The aim of this study was to investigate if Stx signals via ATP and if blockade of purinergic receptors could be protective. Stx induced ATP release from HeLa cells and in a mouse model. Toxin induced rapid calcium influx into HeLa cells, as well as platelets, and a P2X1 receptor antagonist, NF449, abolished this effect. Likewise, the P2X antagonist suramin blocked calcium influx in Hela cells. NF449 did not affect toxin intracellular retrograde transport, however, cells pre-treated with NF449 exhibited significantly higher viability after exposure to Stx for 24 hours, compared to untreated cells. NF449 protected HeLa cells from protein synthesis inhibition and from Stx-induced apoptosis, assayed by caspase 3/7 activity. The latter effect was confirmed by P2X1 receptor silencing. Stx induced the release of toxin-positive HeLa cell- and platelet-derived microvesicles, detected by flow cytometry, an effect significantly reduced by NF449 or suramin. Suramin decreased microvesicle levels in mice injected with Stx or inoculated with Stx-producing EHEC. Taken together, we describe a novel mechanism of Stx-mediated cellular injury associated with ATP signaling and inhibited by P2X receptor blockade.


Subject(s)
Escherichia coli Infections/drug therapy , Hemolytic-Uremic Syndrome/drug therapy , Receptors, Purinergic P2X1/genetics , Shiga Toxin/genetics , Adenosine Triphosphate/metabolism , Animals , Benzenesulfonates/pharmacology , Blood Platelets/microbiology , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , HeLa Cells , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Humans , Mice , Purinergic P2X Receptor Antagonists/pharmacology , Shiga Toxin/antagonists & inhibitors
10.
EBioMedicine ; 47: 319-328, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31444145

ABSTRACT

BACKGROUND: The complement and kallikrein-kinin systems (KKS) are activated during vascular inflammation. The aim of this study was to investigate if blockade of the KKS can affect complement activation on the endothelium during inflammation. METHODS: Complement deposition on endothelial microvesicles was assayed in vasculitis patient plasma samples and controls. Plasma was perfused over glomerular endothelial cells and complement deposition assayed by flow cytometry. The effect of the kinin system was assessed using kinin receptor antagonists and C1-inhibitor. The in vivo effect was assessed in kidney sections from mice with nephrotoxic serum-induced glomerulonephritis treated with a kinin receptor antagonist. FINDINGS: Vasculitis patient plasma had significantly more C3- and C9-positive endothelial microvesicles than controls. Perfusion of patient acute-phase plasma samples over glomerular endothelial cells induced the release of significantly more complement-positive microvesicles, in comparison to remission or control plasma. Complement activation on endothelial microvesicles was reduced by kinin B1- and B2-receptor antagonists or by C1-inhibitor (the main inhibitor of the classical pathway and the KKS). Likewise, perfusion of glomerular endothelial cells with C1-inhibitor-depleted plasma induced the release of complement-positive microvesicles, which was significantly reduced by kinin-receptor antagonists or C1-inhibitor. Mice with nephrotoxic serum-induced glomerulonephritis exhibited significantly reduced glomerular C3 deposition when treated with a B1-receptor antagonist. INTERPRETATION: Excessive complement deposition on the endothelium will promote endothelial injury and the release of endothelial microvesicles. This study demonstrates that blockade of the KKS can reduce complement activation and thereby the inflammatory response on the endothelium. FUNDING: Full details are provided in the Acknowledgements/Funding section.


Subject(s)
Complement Activation/immunology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Kallikrein-Kinin System/drug effects , Vasculitis/etiology , Vasculitis/metabolism , Adult , Aged , Animals , Biological Transport , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Cell-Derived Microparticles/metabolism , Cells, Cultured , Complement C1 Inhibitor Protein/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Models, Animal , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Immunoglobulin G/immunology , Male , Mice , Middle Aged , Protein Binding , Vasculitis/pathology
11.
Kidney Int ; 94(4): 689-700, 2018 10.
Article in English | MEDLINE | ID: mdl-29884545

ABSTRACT

Certain kidney diseases are associated with complement activation although a renal triggering factor has not been identified. Here we demonstrated that renin, a kidney-specific enzyme, cleaves C3 into C3b and C3a, in a manner identical to the C3 convertase. Cleavage was specifically blocked by the renin inhibitor aliskiren. Renin-mediated C3 cleavage and its inhibition by aliskiren also occurred in serum. Generation of C3 cleavage products was demonstrated by immunoblotting, detecting the cleavage product C3b, by N-terminal sequencing of the cleavage product, and by ELISA for C3a release. Functional assays showed mast cell chemotaxis towards the cleavage product C3a and release of factor Ba when the cleavage product C3b was combined with factor B and factor D. The renin-mediated C3 cleavage product bound to factor B. In the presence of aliskiren this did not occur, and less C3 deposited on renin-producing cells. The effect of aliskiren was studied in three patients with dense deposit disease and this demonstrated decreased systemic and renal complement activation (increased C3, decreased C3a and C5a, decreased renal C3 and C5b-9 deposition and/or decreased glomerular basement membrane thickness) over a follow-up period of four to seven years. Thus, renin can trigger complement activation, an effect inhibited by aliskiren. Since renin concentrations are higher in renal tissue than systemically, this may explain the renal propensity of complement-mediated disease in the presence of complement mutations or auto-antibodies.


Subject(s)
Amides/pharmacology , Complement Activation/drug effects , Complement C3/chemistry , Fumarates/pharmacology , Glomerulonephritis, Membranoproliferative/metabolism , Glomerulonephritis, Membranoproliferative/therapy , Renin/chemistry , Amides/therapeutic use , Chemotaxis/drug effects , Child , Complement C3/metabolism , Complement C3a/chemistry , Complement C3a/metabolism , Complement C3b/chemistry , Complement C3b/metabolism , Complement C4/chemistry , Complement C5a/chemistry , Complement C5a/metabolism , Complement C5b/chemistry , Complement C5b/metabolism , Complement Factor B/chemistry , Complement Factor D/chemistry , Female , Fumarates/therapeutic use , Glomerular Basement Membrane/pathology , Glomerulonephritis, Membranoproliferative/pathology , Humans , Mast Cells/physiology , Renin/antagonists & inhibitors , Renin/metabolism
12.
J Am Soc Nephrol ; 28(8): 2472-2481, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28289183

ABSTRACT

The kinin system is activated during vasculitis and may contribute to chronic inflammation. C1-inhibitor is the main inhibitor of the kinin system. In this study, we investigated the presence of the kinin B1 receptor on endothelial microvesicles and its contribution to the inflammatory process. Compared with controls (n=15), patients with acute vasculitis (n=12) had markedly higher levels of circulating endothelial microvesicles, identified by flow cytometry analysis, and significantly more microvesicles that were positive for the kinin B1 receptor (P<0.001). Compared with microvesicles from wild-type cells, B1 receptor-positive microvesicles derived from transfected human embryonic kidney cells induced a significant neutrophil chemotactic effect, and a B1 receptor antagonist blocked this effect. Likewise, patient plasma induced neutrophil chemotaxis, an effect decreased by reduction of microvesicle levels and by blocking the B1 receptor. We used a perfusion system to study the effect of patient plasma (n=6) and control plasma (n=6) on the release of microvesicles from glomerular endothelial cells. Patient samples induced the release of significantly more B1 receptor-positive endothelial microvesicles than control samples, an effect abrogated by reduction of the microvesicles in the perfused samples. Perfusion of C1-inhibitor-depleted plasma over glomerular endothelial cells promoted excessive release of B1 receptor-positive endothelial microvesicles compared with normal plasma, an effect significantly decreased by addition of C1-inhibitor or B1 receptor-antagonist. Thus, B1 receptor-positive endothelial microvesicles may contribute to chronic inflammation by inducing neutrophil chemotaxis, and the reduction of these microvesicles by C1-inhibitor should be explored as a potential treatment for neutrophil-induced inflammation.


Subject(s)
Cell-Derived Microparticles/physiology , Complement C1 Inactivator Proteins/physiology , Vasculitis/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Chemotaxis , Child , Complement C1 Inhibitor Protein , Endothelium, Vascular/cytology , Female , Humans , Male , Middle Aged , Young Adult
13.
EBioMedicine ; 16: 302-311, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28139439

ABSTRACT

Adequate cleavage of von Willebrand factor (VWF) prevents formation of thrombi. ADAMTS13 is the main VWF-cleaving protease and its deficiency results in development of thrombotic microangiopathy. Besides ADAMTS13 other proteases may also possess VWF-cleaving activity, but their physiological importance in preventing thrombus formation is unknown. This study investigated if, and which, proteases could cleave VWF in the glomerulus. The content of the glomerular basement membrane (GBM) was studied as a reflection of processes occurring in the subendothelial glomerular space. VWF was incubated with human GBMs and VWF cleavage was assessed by multimer structure analysis, immunoblotting and mass spectrometry. VWF was cleaved into the smallest multimers by the GBM, which contained ADAMTS13 as well as neutrophil proteases, elastase, proteinase 3 (PR3), cathepsin-G and matrix-metalloproteinase 9. The most potent components of the GBM capable of VWF cleavage were in the serine protease or metalloprotease category, but not ADAMTS13. Neutralization of neutrophil serine proteases inhibited GBM-mediated VWF-cleaving activity, demonstrating a marked contribution of elastase and/or PR3. VWF-platelet strings formed on the surface of primary glomerular endothelial cells, in a perfusion system, were cleaved by both elastase and the GBM, a process blocked by elastase inhibitor. Ultramorphological studies of the human kidney demonstrated neutrophils releasing elastase into the GBM. Neutrophil proteases may contribute to VWF cleavage within the subendothelium, adjacent to the GBM, and thus regulate thrombus size. This anti-thrombotic mechanism would protect the normal kidney during inflammation and could also explain why most patients with ADAMTS13 deficiency do not develop severe kidney failure.


Subject(s)
Kidney Glomerulus/metabolism , Kidney/metabolism , Neutrophils/enzymology , Peptide Hydrolases/metabolism , Thrombosis/metabolism , von Willebrand Factor/metabolism , ADAMTS13 Protein/metabolism , Adult , Blood Platelets/metabolism , Cathepsin G/metabolism , Endothelial Cells/metabolism , Glomerular Basement Membrane/drug effects , Glomerular Basement Membrane/metabolism , Humans , Immunoblotting , Kidney/blood supply , Kidney/ultrastructure , Kidney Glomerulus/drug effects , Kidney Glomerulus/ultrastructure , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Microscopy, Electron, Transmission , Myeloblastin/metabolism , Proteinase Inhibitory Proteins, Secretory/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thrombosis/prevention & control
15.
J Immunol ; 197(4): 1276-86, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27421478

ABSTRACT

Complement activation occurs during enterohemorrhagic Escherichia coli (EHEC) infection and may exacerbate renal manifestations. In this study, we show glomerular C5b-9 deposits in the renal biopsy of a child with EHEC-associated hemolytic uremic syndrome. The role of the terminal complement complex, and its blockade as a therapeutic modality, was investigated in a mouse model of E. coli O157:H7 infection. BALB/c mice were treated with monoclonal anti-C5 i.p. on day 3 or 6 after intragastric inoculation and monitored for clinical signs of disease and weight loss for 14 d. All infected untreated mice (15 of 15) or those treated with an irrelevant Ab (8 of 8) developed severe illness. In contrast, only few infected mice treated with anti-C5 on day 3 developed symptoms (three of eight, p < 0.01 compared with mice treated with the irrelevant Ab on day 3) whereas most mice treated with anti-C5 on day 6 developed symptoms (six of eight). C6-deficient C57BL/6 mice were also inoculated with E. coli O157:H7 and only 1 of 14 developed disease, whereas 10 of 16 wild-type mice developed weight loss and severe disease (p < 0.01). Complement activation via the terminal pathway is thus involved in the development of disease in murine EHEC infection. Early blockade of the terminal complement pathway, before the development of symptoms, was largely protective, whereas late blockade was not. Likewise, lack of C6, and thereby deficient terminal complement complex, was protective in murine E. coli O157:H7 infection.


Subject(s)
Complement C6/antagonists & inhibitors , Complement Membrane Attack Complex/antagonists & inhibitors , Escherichia coli Infections/immunology , Hemolytic-Uremic Syndrome/immunology , Animals , Child, Preschool , Complement C6/immunology , Complement Membrane Attack Complex/immunology , Disease Models, Animal , Enterohemorrhagic Escherichia coli , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
16.
Adv Exp Med Biol ; 865: 19-42, 2015.
Article in English | MEDLINE | ID: mdl-26306441

ABSTRACT

The complement system is activated in the vasculature during thrombotic and inflammatory conditions. Activation may be associated with chronic inflammation on the endothelial surface leading to complement deposition. Complement mutations allow uninhibited complement activation to occur on platelets, neutrophils, monocytes, and aggregates thereof, as well as on red blood cells and endothelial cells. Furthermore, complement activation on the cells leads to the shedding of cell derived-microvesicles that may express complement and tissue factor thus promoting inflammation and thrombosis. Complement deposition on red blood cells triggers hemolysis and the release of red blood cell-derived microvesicles that are prothrombotic. Microvesicles are small membrane vesicles ranging from 0.1 to 1 µm, shed by cells during activation, injury and/or apoptosis that express components of the parent cell. Microvesicles are released during inflammatory and vascular conditions. The repertoire of inflammatory markers on endothelial cell-derived microvesicles shed during inflammation is large and includes complement. These circulating microvesicles may reflect the ongoing inflammatory process but may also contribute to its propagation. This overview will describe complement activation on blood and endothelial cells and the release of microvesicles from these cells during hemolytic uremic syndrome, thrombotic thrombocytopenic purpura and vasculitis, clinical conditions associated with enhanced thrombosis and inflammation.


Subject(s)
Complement Activation , Complement System Proteins/metabolism , Hemolytic-Uremic Syndrome/metabolism , Purpura, Thrombotic Thrombocytopenic/metabolism , Thrombosis/metabolism , Vasculitis/metabolism , Blood Coagulation Factors/immunology , Blood Coagulation Factors/metabolism , Blood Platelets/immunology , Blood Platelets/metabolism , Blood Platelets/pathology , Cell-Derived Microparticles/immunology , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Complement System Proteins/immunology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/pathology , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/pathology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Purpura, Thrombotic Thrombocytopenic/immunology , Purpura, Thrombotic Thrombocytopenic/pathology , Thrombosis/immunology , Thrombosis/pathology , Vasculitis/immunology , Vasculitis/pathology
17.
PLoS Pathog ; 11(2): e1004619, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25719452

ABSTRACT

Shiga toxin (Stx) is the main virulence factor of enterohemorrhagic Escherichia coli, which are non-invasive strains that can lead to hemolytic uremic syndrome (HUS), associated with renal failure and death. Although bacteremia does not occur, bacterial virulence factors gain access to the circulation and are thereafter presumed to cause target organ damage. Stx was previously shown to circulate bound to blood cells but the mechanism by which it would potentially transfer to target organ cells has not been elucidated. Here we show that blood cell-derived microvesicles, shed during HUS, contain Stx and are found within patient renal cortical cells. The finding was reproduced in mice infected with Stx-producing Escherichia coli exhibiting Stx-containing blood cell-derived microvesicles in the circulation that reached the kidney where they were transferred into glomerular and peritubular capillary endothelial cells and further through their basement membranes followed by podocytes and tubular epithelial cells, respectively. In vitro studies demonstrated that blood cell-derived microvesicles containing Stx undergo endocytosis in glomerular endothelial cells leading to cell death secondary to inhibited protein synthesis. This study demonstrates a novel virulence mechanism whereby bacterial toxin is transferred within host blood cell-derived microvesicles in which it may evade the host immune system.


Subject(s)
Bacterial Toxins/metabolism , Blood Cells/metabolism , Cell-Derived Microparticles/metabolism , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Adolescent , Adult , Animals , Blood Cells/microbiology , Cell-Derived Microparticles/microbiology , Cells, Cultured , Child , Child, Preschool , Escherichia coli Infections/pathology , Female , Host-Pathogen Interactions , Humans , Infant , Male , Mice , Mice, Inbred BALB C , Protein Transport
18.
J Immunol ; 194(5): 2309-18, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25637016

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) cause hemolytic uremic syndrome (HUS). This study investigated whether Stx2 induces hemolysis and whether complement is involved in the hemolytic process. RBCs and/or RBC-derived microvesicles from patients with STEC-HUS (n = 25) were investigated for the presence of C3 and C9 by flow cytometry. Patients exhibited increased C3 deposition on RBCs compared with controls (p < 0.001), as well as high levels of C3- and C9-bearing RBC-derived microvesicles during the acute phase, which decreased after recovery. Stx2 bound to P1 (k) and P2 (k) phenotype RBCs, expressing high levels of the P(k) Ag (globotriaosylceramide), the known Stx receptor. Stx2 induced the release of hemoglobin and lactate dehydrogenase in whole blood, indicating hemolysis. Stx2-induced hemolysis was not demonstrated in the absence of plasma and was inhibited by heat inactivation, as well as by the terminal complement pathway Ab eculizumab, the purinergic P2 receptor antagonist suramin, and EDTA. In the presence of whole blood or plasma/serum, Stx2 induced the release of RBC-derived microvesicles coated with C5b-9, a process that was inhibited by EDTA, in the absence of factor B, and by purinergic P2 receptor antagonists. Thus, complement-coated RBC-derived microvesicles are elevated in HUS patients and induced in vitro by incubation of RBCs with Stx2, which also induced hemolysis. The role of complement in Stx2-mediated hemolysis was demonstrated by its occurrence only in the presence of plasma and its abrogation by heat inactivation, EDTA, and eculizumab. Complement activation on RBCs could play a role in the hemolytic process occurring during STEC-HUS.


Subject(s)
Coated Vesicles/drug effects , Erythrocytes/drug effects , Escherichia coli Infections/blood , Escherichia coli O157/pathogenicity , Hemolytic-Uremic Syndrome/blood , Shiga Toxin/toxicity , Adult , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Child , Child, Preschool , Coated Vesicles/chemistry , Coated Vesicles/immunology , Complement Activation/drug effects , Complement C3/chemistry , Complement C9/chemistry , Complement Membrane Attack Complex/chemistry , Edetic Acid/pharmacology , Erythrocytes/chemistry , Erythrocytes/immunology , Erythrocytes/pathology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli O157/immunology , Escherichia coli O157/metabolism , Female , Gene Expression , Hemolysis/drug effects , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Humans , Infant , L-Lactate Dehydrogenase/metabolism , Male , Middle Aged , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/immunology , Shiga Toxin/chemistry , Shiga Toxin/immunology , Suramin/pharmacology , Trihexosylceramides/immunology
19.
J Immunol ; 193(1): 317-26, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24850720

ABSTRACT

IgA nephropathy (IgAN) is characterized by mesangial cell proliferation and extracellular matrix expansion associated with immune deposits consisting of galactose-deficient polymeric IgA1 and C3. We have previously shown that IgA-binding regions of streptococcal M proteins colocalize with IgA in mesangial immune deposits in patients with IgAN. In the present study, the IgA-binding M4 protein from group A Streptococcus was found to bind to galactose-deficient polymeric IgA1 with higher affinity than to other forms of IgA1, as shown by surface plasmon resonance and solid-phase immunoassay. The M4 protein was demonstrated to bind to mesangial cells not via the IgA-binding region but rather via the C-terminal region, as demonstrated by flow cytometry. IgA1 enhanced binding of M4 to mesangial cells, but not vice versa. Costimulation of human mesangial cells with M4 and galactose-deficient polymeric IgA1 resulted in a significant increase in IL-6 secretion compared with each stimulant alone. Galactose-deficient polymeric IgA1 alone, but not M4, induced C3 secretion from the cells, and costimulation enhanced this effect. Additionally, costimulation enhanced mesangial cell proliferation compared with each stimulant alone. These results indicate that IgA-binding M4 protein binds preferentially to galactose-deficient polymeric IgA1 and that these proteins together induce excessive proinflammatory responses and proliferation of human mesangial cells. Thus, tissue deposition of streptococcal IgA-binding M proteins may contribute to the pathogenesis of IgAN.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Complement C3/immunology , Glomerulonephritis, IGA/immunology , Immunoglobulin A/immunology , Interleukin-6/immunology , Mesangial Cells/immunology , Streptococcus/immunology , Adolescent , Female , Glomerulonephritis, IGA/pathology , Humans , Male , Mesangial Cells/pathology , Middle Aged
20.
Nephrol Dial Transplant ; 28(11): 2899-907, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24009284

ABSTRACT

BACKGROUND: Atypical haemolytic uraemic syndrome (aHUS) is associated with dysfunction of the alternative pathway of complement. Disease activity subsides as renal failure progresses but recurs upon renal transplantation, indicating that viable renal tissue contributes to disease activity. We present evidence of cerebrovascular occlusive disease indicating that vascular injury may occur in the absence of kidneys. METHODS: A currently 12-year-old girl developed renal failure at the age of 20 months. She underwent bilateral nephrectomy and renal transplantation but lost the transplant due to recurrences. She was on haemodialysis for 7 years. At 10 years of age she developed a transient ischaemic attack. Imaging, genetic investigation and mutation characterization were performed. RESULTS: Imaging demonstrated occlusion and stenosis of the carotid arteries. Two complement mutations, a novel mutation in factor B and a previously described mutation in factor I, and the H3-factor H haplotype, were identified. The factor B mutation, L433S, did not induce excessive complement activation in vitro. Measurement of C3 degradation products indicated ongoing complement activation. In spite of the patient being anephric, treatment was initiated with eculizumab, a humanized anti-C5 antibody that blocks terminal complement activation. She underwent a successful kidney transplant 9 months later and has not developed a recurrence or progression of vascular stenosis 1 year later. CONCLUSIONS: The course of disease in this patient with aHUS suggests that complement-mediated vascular injury may occur in the total absence of renal tissue and overt recurrences. To our knowledge, this is the first description of eculizumab treatment in an anephric aHUS patient.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Cerebrovascular Disorders/drug therapy , Hemolytic-Uremic Syndrome/drug therapy , Kidney Transplantation , Nephrectomy/adverse effects , Renal Insufficiency/complications , Atypical Hemolytic Uremic Syndrome , Cerebrovascular Disorders/etiology , Cerebrovascular Disorders/pathology , Child , Complement C5/metabolism , Complement Factor B/genetics , Enzyme-Linked Immunosorbent Assay , Female , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/pathology , Humans , Magnetic Resonance Imaging , Mutation/genetics , Polymerase Chain Reaction , Prognosis , Renal Insufficiency/surgery , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...