133 results on '"Diana Karpman"'
Search Results
2. Red blood cell-derived arginase release in hemolytic uremic syndrome
- Author
-
Niklas Friberg, Ida Arvidsson, Ashmita Tontanahal, Ann-Charlotte Kristoffersson, Magnus Gram, Bernard S. Kaplan, and Diana Karpman
- Subjects
Arginase ,Hemolytic uremic syndrome ,Thrombotic microangiopathy ,Nitric oxide ,Shiga toxin ,Medicine - Abstract
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.
- Published
- 2024
- Full Text
- View/download PDF
3. Factor B Mutation in Monozygotic Twins Discordant for Atypical Hemolytic Uremic Syndrome
- Author
-
Sigridur Sunna Aradottir, Ann-Charlotte Kristoffersson, Brynjar O. Jensson, Patrick Sulem, Henning Gong, Runolfur Palsson, and Diana Karpman
- Subjects
atypical hemolytic uremic syndrome ,complement ,Factor B ,genetics ,monozygotic twins ,Diseases of the genitourinary system. Urology ,RC870-923 - Published
- 2023
- Full Text
- View/download PDF
4. Genetic investigation of Nordic patients with complement-mediated kidney diseases
- Author
-
Viktor Rydberg, Sigridur Sunna Aradottir, Ann-Charlotte Kristoffersson, Naila Svitacheva, and Diana Karpman
- Subjects
complement ,atypical hemolytic uremic syndrome ,C3 glomerulopathy ,membranoproliferative glomerulonephritis ,genes ,Immunologic diseases. Allergy ,RC581-607 - Abstract
BackgroundComplement 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.MethodsPatients 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
- Published
- 2023
- Full Text
- View/download PDF
5. Apyrase decreases phage induction and Shiga toxin release from E. coli O157:H7 and has a protective effect during infection
- Author
-
Ida Arvidsson, Ashmita Tontanahal, Karl Johansson, Ann-Charlotte Kristoffersson, Sára Kellnerová, Michael Berger, Ulrich Dobrindt, and Diana Karpman
- Subjects
Enterohemorrhagic Escherichia coli ,RecA ,Shiga toxin ,apyrase ,intestine ,mouse ,Diseases of the digestive system. Gastroenterology ,RC799-869 - 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.
- Published
- 2022
- Full Text
- View/download PDF
6. IgG Binds Escherichia coli Serine Protease EspP and Protects Mice From E. coli O157:H7 Infection
- Author
-
Ashmita Tontanahal, Vanessa Sperandio, Olga Kovbasnjuk, Sebastian Loos, Ann-Charlotte Kristoffersson, Diana Karpman, and Ida Arvidsson
- Subjects
Escherichia coli O157:H7 ,Shiga toxin ,EspP ,immunoglobulin G ,hemolytic uremic syndrome ,mouse ,Immunologic diseases. Allergy ,RC581-607 - 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.
- Published
- 2022
- Full Text
- View/download PDF
7. Factor D Inhibition Blocks Complement Activation Induced by Mutant Factor B Associated With Atypical Hemolytic Uremic Syndrome and Membranoproliferative Glomerulonephritis
- Author
-
Sigridur Sunna Aradottir, Ann-Charlotte Kristoffersson, Lubka T. Roumenina, Anna Bjerre, Pavlos Kashioulis, Runolfur Palsson, and Diana Karpman
- Subjects
complement ,factor B ,factor D ,danicopan ,atypical hemolytic uremic syndrome ,C3 glomerulopathy ,Immunologic diseases. Allergy ,RC581-607 - 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.
- Published
- 2021
- Full Text
- View/download PDF
8. Blockade of the kallikrein-kinin system reduces endothelial complement activation in vascular inflammationResearch in context
- Author
-
Ingrid Lopatko Fagerström, Anne-lie Ståhl, Maria Mossberg, Ramesh Tati, Ann-Charlotte Kristoffersson, Robin Kahn, Jean-Loup Bascands, Julie Klein, Joost P. Schanstra, Mårten Segelmark, and Diana Karpman
- Subjects
Medicine ,Medicine (General) ,R5-920 - 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. Keywords: Vasculitis, Endothelial microvesicles, Complement, Kinin, Kidney, Mouse
- Published
- 2019
- Full Text
- View/download PDF
9. Shiga Toxin-Bearing Microvesicles Exert a Cytotoxic Effect on Recipient Cells Only When the Cells Express the Toxin Receptor
- Author
-
Karl Johansson, Annie Willysson, Ann-Charlotte Kristoffersson, Ashmita Tontanahal, Daniel Gillet, Anne-lie Ståhl, and Diana Karpman
- Subjects
Shiga toxin ,enterohemorrhagic Escherichia coli ,hemolytic uremic syndrome ,microvesicles ,globotriaosylceramide ,Gb3 ,Microbiology ,QR1-502 - 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.
- Published
- 2020
- Full Text
- View/download PDF
10. Neutrophil Protease Cleavage of Von Willebrand Factor in Glomeruli – An Anti-thrombotic Mechanism in the Kidney
- Author
-
Ramesh Tati, Ann-Charlotte Kristoffersson, Minola Manea Hedström, Matthias Mörgelin, Jörgen Wieslander, Cees van Kooten, and Diana Karpman
- Subjects
Von Willebrand factor ,Glomerular basement membrane ,Kidney ,Neutrophils ,ADAMTS13 ,Elastase ,Medicine ,Medicine (General) ,R5-920 - 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.
- Published
- 2017
- Full Text
- View/download PDF
11. Annexin Induces Cellular Uptake of Extracellular Vesicles and Delays Disease in Escherichia coli O157:H7 Infection
- Author
-
Ashmita Tontanahal, Ida Arvidsson, and Diana Karpman
- Subjects
Annexin A5 ,extracellular vesicles ,enterohemorrhagic Escherichia coli ,Shiga toxin ,hemolytic uremic syndrome ,phagocytes ,Biology (General) ,QH301-705.5 - Abstract
Enterohemorrhagic Escherichia coli secrete Shiga toxin and lead to hemolytic uremic syndrome. Patients have high levels of circulating prothrombotic extracellular vesicles (EVs) that expose phosphatidylserine and tissue factor and transfer Shiga toxin from the circulation into the kidney. Annexin A5 (AnxA5) binds to phosphatidylserine, affecting membrane dynamics. This study investigated the effect of anxA5 on EV uptake by human and murine phagocytes and used a mouse model of EHEC infection to study the effect of anxA5 on disease and systemic EV levels. EVs derived from human whole blood or HeLa cells were more readily taken up by THP-1 cells or RAW264.7 cells when the EVs were coated with anxA5. EVs from HeLa cells incubated with RAW264.7 cells induced phosphatidylserine exposure on the cells, suggesting a mechanism by which anxA5-coated EVs can bind to phagocytes before uptake. Mice treated with anxA5 for six days after inoculation with E. coli O157:H7 showed a dose-dependent delay in the development of clinical disease. Treated mice had lower levels of EVs in the circulation. In the presence of anxA5, EVs are taken up by phagocytes and their systemic levels are lower, and, as EVs transfer Shiga toxin to the kidney, this could postpone disease development.
- Published
- 2021
- Full Text
- View/download PDF
12. Shiga Toxin Uptake and Sequestration in Extracellular Vesicles Is Mediated by Its B-Subunit
- Author
-
Annie Willysson, Anne-lie Ståhl, Daniel Gillet, Julien Barbier, Jean-Christophe Cintrat, Valérie Chambon, Anne Billet, Ludger Johannes, and Diana Karpman
- Subjects
Shiga toxin ,extracellular vesicles ,red blood cells ,HeLa cells ,globotriaosylceramide ,Medicine - Abstract
Shiga toxin (Stx)-stimulated blood cells shed extracellular vesicles (EVs) which can transfer the toxin to the kidneys and lead to hemolytic uremic syndrome. The toxin can be taken up by renal cells within EVs wherein the toxin is released, ultimately leading to cell death. The mechanism by which Stx is taken up, translocated, and sequestered in EVs was addressed in this study utilizing the B-subunit that binds to the globotriaosylceramide (Gb3) receptor. We found that Stx1B was released in EVs within minutes after stimulation of HeLa cells or red blood cells, detected by live cell imaging and flow cytometry. In the presence of Retro-2.1, an inhibitor of intracellular retrograde trafficking, a continuous release of Stx-positive EVs occurred. EVs from HeLa cells possess the Gb3 receptor on their membrane, and EVs from cells that were treated with a glycosylceramide synthase inhibitor, to reduce Gb3, bound significantly less Stx1B. Stx1B was detected both on the membrane and within the shed EVs. Stx1B was incubated with EVs derived from blood cells, in the absence of cells, and was shown to bind to, and be taken up by, these EVs, as demonstrated by electron microscopy. Using a membrane translocation assay we demonstrated that Stx1B was taken up by blood cell- and HeLa-derived EVs, an effect enhanced by chloropromazine or methyl-ß-cyclodextrin, suggesting toxin transfer within the membrane. This is a novel mechanism by which EVs derived from blood cells can sequester their toxic content, possibly to evade the host response.
- Published
- 2020
- Full Text
- View/download PDF
13. Clinical and Complement Long-Term Follow-Up of a Pediatric Patient with C3 Mutation-Related Atypical Hemolytic Uremic Syndrome
- Author
-
Anna Bjerre, Grethe Bergseth, Judith Krey Ludviksen, Arne Stokke, Vidar Bosnes, Diana Karpman, and Tom Eirik Mollnes
- Subjects
Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
We report a pediatric patient with atypical hemolytic uremic syndrome due to a C3 gain-of-function mutation diagnosed in infancy. She was treated from the start with a constant dose of 300 mg eculizumab every second week from the onset and followed by routine complement analyses for six years. Her complement system was completely inhibited and the dose interval was prolonged from 2 to 3 weeks without alteration of the dose and the complement activity continued to be completely inhibited. Blood samples taken immediately before, immediately after, and between eculizumab doses were analyzed for eculizumab-C5 complexes and percentage of total complement activity, using the Wieslab® test, and compared to a pool of sera from 20 healthy controls. The patient exhibited complete complement inhibition at all three time-points and had no free circulating C5 suggesting there was complete binding to eculizumab. She has now been treated for six years with full complement blockade. We suggest therefore that analysis of complement activity using the Wieslab® test is useful for evaluating the effect of eculizumab when dose intervals are prolonged.
- Published
- 2018
- Full Text
- View/download PDF
14. A novel mechanism of bacterial toxin transfer within host blood cell-derived microvesicles.
- Author
-
Anne-lie Ståhl, Ida Arvidsson, Karl E Johansson, Milan Chromek, Johan Rebetz, Sebastian Loos, Ann-Charlotte Kristoffersson, Zivile D Békássy, Matthias Mörgelin, and Diana Karpman
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - 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.
- Published
- 2015
- Full Text
- View/download PDF
15. Microvesicle Involvement in Shiga Toxin-Associated Infection
- Author
-
Annie Villysson, Ashmita Tontanahal, and Diana Karpman
- Subjects
Shiga toxin ,hemolytic uremic syndrome ,enterohemorrhagic Escherichia coli ,microvesicles ,kidney ,Medicine - Abstract
Shiga toxin is the main virulence factor of enterohemorrhagic Escherichia coli, a non-invasive pathogen that releases virulence factors in the intestine, causing hemorrhagic colitis and, in severe cases, hemolytic uremic syndrome (HUS). HUS manifests with acute renal failure, hemolytic anemia and thrombocytopenia. Shiga toxin induces endothelial cell damage leading to platelet deposition in thrombi within the microvasculature and the development of thrombotic microangiopathy, mostly affecting the kidney. Red blood cells are destroyed in the occlusive capillary lesions. This review focuses on the importance of microvesicles shed from blood cells and their participation in the prothrombotic lesion, in hemolysis and in the transfer of toxin from the circulation into the kidney. Shiga toxin binds to blood cells and may undergo endocytosis and be released within microvesicles. Microvesicles normally contribute to intracellular communication and remove unwanted components from cells. Many microvesicles are prothrombotic as they are tissue factor- and phosphatidylserine-positive. Shiga toxin induces complement-mediated hemolysis and the release of complement-coated red blood cell-derived microvesicles. Toxin was demonstrated within blood cell-derived microvesicles that transported it to renal cells, where microvesicles were taken up and released their contents. Microvesicles are thereby involved in all cardinal aspects of Shiga toxin-associated HUS, thrombosis, hemolysis and renal failure.
- Published
- 2017
- Full Text
- View/download PDF
16. The antimicrobial peptide cathelicidin protects mice from Escherichia coli O157:H7-mediated disease.
- Author
-
Milan Chromek, Ida Arvidsson, and Diana Karpman
- Subjects
Medicine ,Science - Abstract
This study investigated the role of the antimicrobial peptide cathelicidin in Escherichia coli O157:H7 infection and subsequent renal damage. Mouse and human cathelicidin, CRAMP and LL-37, respectively, killed E. coli O157:H7 in vitro. Intestines from healthy wild-type (129/SvJ) and cathelicidin-knock-out (Camp(-/-)) mice were investigated, showing that cathelicidin-deficient mice had a thinner colonic mucus layer compared with wild-type mice. Wild-type (n = 11) and cathelicidin-knock-out (n = 11) mice were inoculated with E. coli O157:H7. Cathelicidin-deficient animals exhibited higher fecal counts of E. coli O157:H7 and bacteria penetrated the mucus forming attaching-and-effacing lesions to a much higher extent than in wild-type animals. Cathelicidin knock-out mice developed symptoms (9/11) as well as anemia, thrombocytopenia and extensive renal tubular damage while all cathelicidin-producing mice remained asymptomatic with normal laboratory findings. When injected with Shiga toxin intraperitoneally, both murine strains developed the same degree of renal tubular damage and clinical disease indicating that differences in sensitivity to infection between the murine strains were related to the initial intestinal response. In conclusion, cathelicidin substantially influenced the antimicrobial barrier in the mouse colon mucosa. Cathelicidin deficiency lead to increased susceptibility to E. coli O157:H7 infection and subsequent renal damage. Administration of cathelicidin or stimulation of endogenous production may prove to be novel treatments for E. coli O157:H7-induced hemolytic uremic syndrome.
- Published
- 2012
- Full Text
- View/download PDF
17. Phenotypic expression of ADAMTS13 in glomerular endothelial cells.
- Author
-
Ramesh Tati, Ann-Charlotte Kristoffersson, Anne-Lie Ståhl, Matthias Mörgelin, David Motto, Simon Satchell, Peter Mathieson, Minola Manea-Hedström, and Diana Karpman
- Subjects
Medicine ,Science - Abstract
ADAMTS13 is the physiological von Willebrand factor (VWF)-cleaving protease. The aim of this study was to examine ADAMTS13 expression in kidneys from ADAMTS13 wild-type (Adamts13⁺/⁺) and deficient (Adamts13⁻/⁻) mice and to investigate the expression pattern and bioactivity in human glomerular endothelial cells.Immunohistochemistry was performed on kidney sections from ADAMTS13 wild-type and ADAMTS13-deficient mice. Phenotypic differences were examined by ultramorphology. ADAMTS13 expression in human glomerular endothelial cells and dermal microvascular endothelial cells was investigated by real-time PCR, flow cytometry, immunofluorescence and immunoblotting. VWF cleavage was demonstrated by multimer structure analysis and immunoblotting. ADAMTS13 was demonstrated in glomerular endothelial cells in Adamts13⁺/⁺ mice but no staining was visible in tissue from Adamts13⁻/⁻ mice. Thickening of glomerular capillaries with platelet deposition on the vessel wall was detected in Adamts13⁻/⁻ mice. ADAMTS13 mRNA and protein were detected in both human endothelial cells and the protease was secreted. ADAMTS13 activity was demonstrated in glomerular endothelial cells as cleavage of VWF.Glomerular endothelial cells express and secrete ADAMTS13. The proteolytic activity could have a protective effect preventing deposition of platelets along capillary lumina under the conditions of high shear stress present in glomerular capillaries.
- Published
- 2011
- Full Text
- View/download PDF
18. Pathogen specific, IRF3-dependent signaling and innate resistance to human kidney infection.
- Author
-
Hans Fischer, Nataliya Lutay, Bryndís Ragnarsdóttir, Manisha Yadav, Klas Jönsson, Alexander Urbano, Ahmed Al Hadad, Sebastian Rämisch, Petter Storm, Ulrich Dobrindt, Ellaine Salvador, Diana Karpman, Ulf Jodal, and Catharina Svanborg
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The mucosal immune system identifies and fights invading pathogens, while allowing non-pathogenic organisms to persist. Mechanisms of pathogen/non-pathogen discrimination are poorly understood, as is the contribution of human genetic variation in disease susceptibility. We describe here a new, IRF3-dependent signaling pathway that is critical for distinguishing pathogens from normal flora at the mucosal barrier. Following uropathogenic E. coli infection, Irf3(-/-) mice showed a pathogen-specific increase in acute mortality, bacterial burden, abscess formation and renal damage compared to wild type mice. TLR4 signaling was initiated after ceramide release from glycosphingolipid receptors, through TRAM, CREB, Fos and Jun phosphorylation and p38 MAPK-dependent mechanisms, resulting in nuclear translocation of IRF3 and activation of IRF3/IFNβ-dependent antibacterial effector mechanisms. This TLR4/IRF3 pathway of pathogen discrimination was activated by ceramide and by P-fimbriated E. coli, which use ceramide-anchored glycosphingolipid receptors. Relevance of this pathway for human disease was supported by polymorphic IRF3 promoter sequences, differing between children with severe, symptomatic kidney infection and children who were asymptomatic bacterial carriers. IRF3 promoter activity was reduced by the disease-associated genotype, consistent with the pathology in Irf3(-/-) mice. Host susceptibility to common infections like UTI may thus be strongly influenced by single gene modifications affecting the innate immune response.
- Published
- 2010
- Full Text
- View/download PDF
19. Toll-like receptor 4 promoter polymorphisms: common TLR4 variants may protect against severe urinary tract infection.
- Author
-
Bryndís Ragnarsdóttir, Klas Jönsson, Alexander Urbano, Jenny Grönberg-Hernandez, Nataliya Lutay, Martti Tammi, Mattias Gustafsson, Ann-Charlotte Lundstedt, Irene Leijonhufvud, Diana Karpman, Björn Wullt, Lennart Truedsson, Ulf Jodal, Björn Andersson, and Catharina Svanborg
- Subjects
Medicine ,Science - Abstract
BackgroundPolymorphisms affecting Toll-like receptor (TLR) structure appear to be rare, as would be expected due to their essential coordinator role in innate immunity. Here, we assess variation in TLR4 expression, rather than structure, as a mechanism to diversify innate immune responses.Methodology/principal findingsWe sequenced the TLR4 promoter (4,3 kb) in Swedish blood donors. Since TLR4 plays a vital role in susceptibility to urinary tract infection (UTI), promoter sequences were obtained from children with mild or severe disease. We performed a case-control study of pediatric patients with asymptomatic bacteriuria (ABU) or those prone to recurrent acute pyelonephritis (APN). Promoter activity of the single SNPs or multiple allelic changes corresponding to the genotype patterns (GPs) was tested. We then conducted a replication study in an independent cohort of adult patients with a history of childhood APN. Last, in vivo effects of the different GPs were examined after therapeutic intravesical inoculation of 19 patients with Escherichia coli 83972. We identified in total eight TLR4 promoter sequence variants in the Swedish control population, forming 19 haplotypes and 29 genotype patterns, some with effects on promoter activity. Compared to symptomatic patients and healthy controls, ABU patients had fewer genotype patterns, and their promoter sequence variants reduced TLR4 expression in response to infection. The ABU associated GPs also reduced innate immune responses in patients who were subjected to therapeutic urinary E. coli tract inoculation.ConclusionsThe results suggest that genetic variation in the TLR4 promoter may be an essential, largely overlooked mechanism to influence TLR4 expression and UTI susceptibility.
- Published
- 2010
- Full Text
- View/download PDF
20. Shiga toxin and lipopolysaccharide induce platelet-leukocyte aggregates and tissue factor release, a thrombotic mechanism in hemolytic uremic syndrome.
- Author
-
Anne-lie Ståhl, Lisa Sartz, Anders Nelsson, Zivile D Békássy, and Diana Karpman
- Subjects
Medicine ,Science - Abstract
BACKGROUND:Aggregates formed between leukocytes and platelets in the circulation lead to release of tissue factor (TF)-bearing microparticles contributing to a prothrombotic state. As enterohemorrhagic Escherichia coli (EHEC) may cause hemolytic uremic syndrome (HUS), in which microthrombi cause tissue damage, this study investigated whether the interaction between blood cells and EHEC virulence factors Shiga toxin (Stx) and lipopolysaccharide (LPS) led to release of TF. METHODOLOGY/PRINCIPAL FINDINGS:The interaction between Stx or LPS and blood cells induced platelet-leukocyte aggregate formation and tissue factor (TF) release, as detected by flow cytometry in whole blood. O157LPS was more potent than other LPS serotypes. Aggregates formed mainly between monocytes and platelets and less so between neutrophils and platelets. Stimulated blood cells in complex expressed activation markers, and microparticles were released. Microparticles originated mainly from platelets and monocytes and expressed TF. TF-expressing microparticles, and functional TF in plasma, increased when blood cells were simultaneously exposed to the EHEC virulence factors and high shear stress. Stx and LPS in combination had a more pronounced effect on platelet-monocyte aggregate formation, and TF expression on these aggregates, than each virulence factor alone. Whole blood and plasma from HUS patients (n = 4) were analyzed. All patients had an increase in leukocyte-platelet aggregates, mainly between monocytes and platelets, on which TF was expressed during the acute phase of disease. Patients also exhibited an increase in microparticles, mainly originating from platelets and monocytes, bearing surface-bound TF, and functional TF was detected in their plasma. Blood cell aggregates, microparticles, and TF decreased upon recovery. CONCLUSIONS/SIGNIFICANCE:By triggering TF release in the circulation, Stx and LPS can induce a prothrombotic state contributing to the pathogenesis of HUS.
- Published
- 2009
- Full Text
- View/download PDF
21. A genetic basis of susceptibility to acute pyelonephritis.
- Author
-
Ann-Charlotte Lundstedt, Shane McCarthy, Mattias C U Gustafsson, Gabriela Godaly, Ulf Jodal, Diana Karpman, Irene Leijonhufvud, Carin Lindén, Jeanette Martinell, Bryndis Ragnarsdottir, Martin Samuelsson, Lennart Truedsson, Björn Andersson, and Catharina Svanborg
- Subjects
Medicine ,Science - Abstract
BackgroundFor unknown reasons, urinary tract infections (UTIs) are clustered in certain individuals. Here we propose a novel, genetically determined cause of susceptibility to acute pyelonephritis, which is the most severe form of UTI. The IL-8 receptor, CXCR1, was identified as a candidate gene when mIL-8Rh mutant mice developed acute pyelonephritis (APN) with severe tissue damage.Methods and findingsWe have obtained CXCR1 sequences from two, highly selected APN prone patient groups, and detected three unique mutations and two known polymorphisms with a genotype frequency of 23% and 25% compared to 7% in controls (pConclusionsThe results identify a genetic innate immune deficiency, with a strong link to APN and renal scarring.
- Published
- 2007
- Full Text
- View/download PDF
22. A link between Krüppel-like factor 4, complement activation, and kidney damage
- Author
-
Sebastian Loos and Diana Karpman
- Subjects
Kruppel-Like Factor 4 ,Mice ,Thrombotic Microangiopathies ,Nephrology ,Kidney Glomerulus ,Animals ,Humans ,Kidney Diseases ,Complement Activation ,Transcription Factors - Abstract
Krüppel-like factors (KLFs) are transcription factors with important roles in tissue homeostasis. KLF4 possesses antithrombotic and anti-inflammatory properties. In this issue, Estrada et al. show that endothelial KLF4 prevents complement deposition in glomeruli and in its absence the cell-bound complement regulator CD55 was reduced. The study included endothelial-specific KLF4 knockdown mice that mimic thrombotic microangiopathy and thrombotic microangiopathy patient biopsies showing decreased KLF4 and CD55. The results suggest that KLF4 is involved in the regulation of glomerular complement deposition.
- Published
- 2022
- Full Text
- View/download PDF
23. Crosstalk between the renin–angiotensin, complement and kallikrein–kinin systems in inflammation
- Author
-
Ingrid Lopatko Fagerström, Diana Karpman, Michael Bader, and Zivile Békássy
- Subjects
History ,Kallikrein-Kinin System ,Inflammation ,Review Article ,Inflammatory diseases ,Systemic inflammation ,Education ,Renin-Angiotensin System ,Renin–angiotensin system ,Humans ,Medicine ,business.industry ,COVID-19 ,Thrombosis ,Chemotaxis ,Complement System Proteins ,medicine.disease ,Computer Science Applications ,Complement system ,Complement cascade ,Crosstalk (biology) ,Coagulation ,Hereditary angioedema ,Immunology ,medicine.symptom ,business - Abstract
During severe inflammatory and infectious diseases, various mediators modulate the equilibrium of vascular tone, inflammation, coagulation and thrombosis. This Review describes the interactive roles of the renin–angiotensin system, the complement system, and the closely linked kallikrein–kinin and contact systems in cell biological functions such as vascular tone and leakage, inflammation, chemotaxis, thrombosis and cell proliferation. Specific attention is given to the role of these systems in systemic inflammation in the vasculature and tissues during hereditary angioedema, cardiovascular and renal glomerular disease, vasculitides and COVID-19. Moreover, we discuss the therapeutic implications of these complex interactions, given that modulation of one system may affect the other systems, with beneficial or deleterious consequences., The renin–angiotensin, complement and kallikrein–kinin systems comprise a multitude of mediators that modulate physiological responses during inflammatory and infectious diseases. This Review investigates the complex interactions between these systems and how these are dysregulated in various conditions, including cardiovascular diseases and COVID-19, as well as their therapeutic implications.
- Published
- 2021
- Full Text
- View/download PDF
24. In Vivo Sustained Release of the Retrograde Transport Inhibitor Retro-2.1 Formulated in a Thermosensitive Hydrogel
- Author
-
Robin Vinck, Laetitia Anvi Nguyen, Mathilde Munier, Lucie Caramelle, Diana Karpman, Julien Barbier, Alain Pruvost, Jean-Christophe Cintrat, and Daniel Gillet
- Subjects
Polymers ,Organic Chemistry ,Temperature ,Hydrogels ,General Medicine ,Catalysis ,retrograde transport inhibitor ,broad spectrum ,Retro-2.1 ,formulation ,thermosensitive hydrogel ,pharmacokinetic ,metabolism ,Polyethylene Glycols ,Computer Science Applications ,Inorganic Chemistry ,Mice ,Delayed-Action Preparations ,Animals ,Nanoparticles ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy - Abstract
A recently developed inhibitor of retrograde transport, namely Retro-2.1, proved to be a potent and broad-spectrum lead in vitro against intracellular pathogens, such as toxins, parasites, intracellular bacteria and viruses. To circumvent its low aqueous solubility, a formulation in poly(ethylene glycol)-block-poly(D,L)lactide micelle nanoparticles was developed. This formulation enabled the study of the pharmacokinetic parameters of Retro-2.1 in mice following intravenous and intraperitoneal injections, revealing a short blood circulation time, with an elimination half-life of 5 and 6.7 h, respectively. To explain the poor pharmacokinetic parameters, the metabolic stability of Retro-2.1 was studied in vitro and in vivo, revealing fast cytochrome-P-450-mediated metabolism into a less potent hydroxylated analogue. Subcutaneous injection of Retro-2.1 formulated in a biocompatible and bioresorbable polymer-based thermosensitive hydrogel allowed for sustained release of the drug, with an elimination half-life of 19 h, and better control of its metabolism. This study provides a guideline on how to administer this promising lead in vivo in order to study its efficacy.
- Published
- 2022
- Full Text
- View/download PDF
25. Extracellular vesicles in renal inflammatory and infectious diseases
- Author
-
Ashmita Tontanahal and Diana Karpman
- Subjects
0301 basic medicine ,Chemokine ,Lupus nephritis ,Inflammation ,Kidney ,Communicable Diseases ,Biochemistry ,Nephropathy ,Extracellular Vesicles ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Physiology (medical) ,medicine ,Humans ,Nephritis ,biology ,business.industry ,Acute kidney injury ,Glomerulonephritis ,medicine.disease ,Kidney Tubules ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Cancer research ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Extracellular vesicles can mediate cell-to-cell communication, or relieve the parent cell of harmful substances, in order to maintain cellular integrity. The content of extracellular vesicles includes miRNAs, mRNAs, growth factors, complement factors, cytokines, chemokines and receptors. These may contribute to inflammatory and infectious diseases by the exposure or transfer of potent effectors that induce vascular inflammation by leukocyte recruitment and thrombosis. Furthermore, vesicles release cytokines and induce their release from cells. Extracellular vesicles possess immune modulatory and anti-microbial properties, and induce receptor signaling in the recipient cell, not least by the transfer of pro-inflammatory receptors. Additionally, the vesicles may carry virulence factors systemically. Extracellular vesicles in blood and urine can contribute to the development of kidney diseases or exhibit protective effects. In this review we will describe the role of EVs in inflammation, thrombosis, immune modulation, angiogenesis, oxidative stress, renal tubular regeneration and infection. Furthermore, we will delineate their contribution to renal ischemia/reperfusion, vasculitis, glomerulonephritis, lupus nephritis, thrombotic microangiopathies, IgA nephropathy, acute kidney injury, urinary tract infections and renal transplantation. Due to their content of miRNAs and growth factors, or when loaded with nephroprotective modulators, extracellular vesicles have the potential to be used as therapeutics for renal regeneration.
- Published
- 2021
- Full Text
- View/download PDF
26. A role for complement blockade in kidney transplantation
- Author
-
Diana Karpman, Zivile Bekassy, Anne Grunenwald, and Lubka T. Roumenina
- Subjects
Graft Rejection ,Complement cascade ,Infectious Diseases ,Preclinical research ,Immunology ,Allotransplantation ,Immunology and Allergy ,Complement System Proteins ,Kidney ,Kidney Transplantation ,Article ,Immunosuppression - Abstract
Sensitized kidney transplant recipients experience high rates of antibody-mediated rejection due to the presence of donor-specific antibodies and immunologic memory. Here we show that transient peri-transplant treatment with the central complement component C3 inhibitor Cp40 significantly prolongs median allograft survival in a sensitized nonhuman primate model. Despite donor-specific antibody levels remaining high, fifty percent of Cp40-treated primates maintain normal kidney function beyond the last day of treatment. Interestingly, presence of antibodies of the IgM class associates with reduced median graft survival (8 vs. 40 days; p = 0.02). Cp40 does not alter lymphocyte depletion by rhesus-specific anti-thymocyte globulin, but inhibits lymphocyte activation and proliferation, resulting in reduced antibody-mediated injury and complement deposition. In summary, Cp40 prevents acute antibody-mediated rejection and prolongs graft survival in primates, and inhibits T and B cell activation and proliferation, suggesting an immunomodulatory effect beyond its direct impact on antibody-mediated injury., Donor-specific antibodies in sensitized recipients may cause kidney transplant rejection. Here the authors show that complement component C3 inhibition prolongs graft survival by inhibiting T and B cell proliferation/activation and hence tissue injury, despite antibody levels remaining unaffected.
- Published
- 2022
27. IgG Binds
- Author
-
Ashmita, Tontanahal, Vanessa, Sperandio, Olga, Kovbasnjuk, Sebastian, Loos, Ann-Charlotte, Kristoffersson, Diana, Karpman, and Ida, Arvidsson
- Subjects
Mice ,Escherichia coli Proteins ,Immunoglobulin G ,Serine Endopeptidases ,Serine ,Animals ,Serine Proteases ,Escherichia coli O157 ,Escherichia coli Infections - Abstract
Shiga toxin-producing
- Published
- 2021
28. Isolation and Characterization of Shiga Toxin-Associated Microvesicles
- Author
-
Diana Karpman, Anne-lie Ståhl, and Annie Willysson
- Subjects
0301 basic medicine ,biology ,Cell ,Shiga toxin ,030204 cardiovascular system & hematology ,medicine.disease ,medicine.disease_cause ,Microvesicles ,Hemolysis ,Virulence factor ,Microbiology ,Blood cell ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cell culture ,medicine ,biology.protein ,Escherichia coli - Abstract
Microvesicles are shed from cell surfaces during infectious or inflammatory conditions and may contribute to the pathogenesis of disease. During Shiga toxin-producing Escherichia coli (STEC) infection, microvesicles are released from blood cells. These microvesicles play a part in inflammation, thrombosis, hemolysis, and the transfer of the main virulence factor of STEC strains, Shiga toxin, to target organ cells. This chapter describes how to isolate blood cell- and cell culture-derived microvesicles from plasma or cell culture medium, respectively, and how to characterize these microvesicles by various methods, with special focus on Shiga toxin-associated microvesicles.
- Published
- 2021
- Full Text
- View/download PDF
29. Exosomes and microvesicles in normal physiology, pathophysiology, and renal diseases
- Author
-
Diana Karpman, Karl Johansson, Robin Kahn, Maria Mossberg, and Anne-lie Ståhl
- Subjects
Senescence ,Nephrology ,medicine.medical_specialty ,Angiogenesis ,030232 urology & nephrology ,Physiology ,Inflammation ,Apoptosis ,Review ,Cell Communication ,030204 cardiovascular system & hematology ,Exosomes ,Kidney ,03 medical and health sciences ,0302 clinical medicine ,Cell-Derived Microparticles ,Internal medicine ,Medicine ,Humans ,Cellular Senescence ,business.industry ,Thrombosis ,Extracellular vesicles ,Pathophysiology ,Microvesicles ,Cell biology ,medicine.anatomical_structure ,Pediatrics, Perinatology and Child Health ,Kidney Diseases ,medicine.symptom ,business ,Intracellular - Abstract
Extracellular vesicles are cell-derived membrane particles ranging from 30 to 5,000 nm in size, including exosomes, microvesicles, and apoptotic bodies. They are released under physiological conditions, but also upon cellular activation, senescence, and apoptosis. They play an important role in intercellular communication. Their release may also maintain cellular integrity by ridding the cell of damaging substances. This review describes the biogenesis, uptake, and detection of extracellular vesicles in addition to the impact that they have on recipient cells, focusing on mechanisms important in the pathophysiology of kidney diseases, such as thrombosis, angiogenesis, tissue regeneration, immune modulation, and inflammation. In kidney diseases, extracellular vesicles may be utilized as biomarkers, as they are detected in both blood and urine. Furthermore, they may contribute to the pathophysiology of renal disease while also having beneficial effects associated with tissue repair. Because of their role in the promotion of thrombosis, inflammation, and immune-mediated disease, they could be the target of drug therapy, whereas their favorable effects could be utilized therapeutically in acute and chronic kidney injury.
- Published
- 2017
30. Neutrophil Protease Cleavage of Von Willebrand Factor in Glomeruli – An Anti-thrombotic Mechanism in the Kidney
- Author
-
Minola Manea Hedström, Jörgen Wieslander, Cees van Kooten, Ann-Charlotte Kristoffersson, Ramesh Tati, Matthias Mörgelin, and Diana Karpman
- Subjects
Male ,0301 basic medicine ,Cathepsin G ,Neutrophils ,Kidney Glomerulus ,lcsh:Medicine ,Review Article ,Von Willebrand factor ,030204 cardiovascular system & hematology ,Kidney ,0302 clinical medicine ,Proteinase 3 ,Elastase ,hemic and lymphatic diseases ,lcsh:R5-920 ,biology ,Chemistry ,Glomerular basement membrane ,General Medicine ,ADAMTS13 ,Elastase inhibitor ,medicine.anatomical_structure ,Matrix Metalloproteinase 9 ,Biochemistry ,cardiovascular system ,lcsh:Medicine (General) ,VonWillebrand factor ,circulatory and respiratory physiology ,Adult ,Blood Platelets ,Proteases ,Myeloblastin ,Immunoblotting ,Proteinase Inhibitory Proteins, Secretory ,ADAMTS13 Protein ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Microscopy, Electron, Transmission ,medicine ,Humans ,Serine protease ,urogenital system ,lcsh:R ,Endothelial Cells ,Thrombosis ,Molecular biology ,030104 developmental biology ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,biology.protein ,Leukocyte Elastase ,Peptide Hydrolases - 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., Highlights • Neutrophil proteases in the glomerular basement membrane cleave VWF and may protect the kidney from microthrombi. • VWF cleavage would be activated by neutrophil influx and compensate for the prothrombotic mechanisms during inflammation. • This mechanism may compensate for lack of ADAMTS13 and explain why TTP patients seldom develop end-stage renal failure. The study demonstrates a mechanism by which the kidney is protected from blood clotting during inflammation. In the inflammatory setting white blood cells infiltrate tissues. In this study we showed that enzymes released from white blood cells into the kidney decrease the size of blood clots. This is a general mechanism but could also explain why patients with thrombotic thrombocytopenic purpura, who develop widespread blood clots in many organs, do not usually develop severe kidney failure.
- Published
- 2017
- Full Text
- View/download PDF
31. Shiga toxin signals via ATP and its effect is blocked by purinergic receptor antagonism
- Author
-
Ida Arvidsson, Karl Johansson, Anne-lie Ståhl, Milan Chromek, Sebastian Loos, Ann-Charlotte Kristoffersson, Diana Karpman, Johan Rebetz, Ashmita Tontanahal, and Ludger Johannes
- Subjects
Blood Platelets ,0301 basic medicine ,Purinergic P2X Receptor Antagonists ,Suramin ,lcsh:Medicine ,Caspase 3 ,Article ,Shiga Toxin ,HeLa ,Mice ,03 medical and health sciences ,Adenosine Triphosphate ,Haemolytic uraemic syndrome ,medicine ,Animals ,Humans ,lcsh:Science ,Receptor ,Escherichia coli Infections ,Multidisciplinary ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Microvesicle ,lcsh:R ,Benzenesulfonates ,Purinergic receptor ,Bacteriology ,Shiga toxin ,biology.organism_classification ,Molecular biology ,Receptors, Purinergic P2X1 ,030104 developmental biology ,Apoptosis ,Enterohemorrhagic Escherichia coli ,Hemolytic-Uremic Syndrome ,biology.protein ,lcsh:Q ,HeLa Cells ,medicine.drug - 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.
- Published
- 2019
- Full Text
- View/download PDF
32. Orphan drug policies and use in pediatric nephrology
- Author
-
Peter Höglund and Diana Karpman
- Subjects
Drug ,medicine.medical_specialty ,Pediatrics ,Drug Industry ,Orphan Drug Production ,Cost-Benefit Analysis ,media_common.quotation_subject ,030232 urology & nephrology ,Psychological intervention ,Drug Costs ,Orphan drug ,03 medical and health sciences ,Rare Diseases ,0302 clinical medicine ,Atypical hemolytic uremic syndrome ,medicine ,Humans ,030212 general & internal medicine ,Child ,Intensive care medicine ,health care economics and organizations ,Reimbursement ,media_common ,business.industry ,Eculizumab ,medicine.disease ,Quality-adjusted life year ,Policy ,Nephrology ,Pediatrics, Perinatology and Child Health ,Kidney Diseases ,business ,medicine.drug ,Rare disease - Abstract
Orphan drugs designed to treat rare diseases are often overpriced per patient. Novel treatments are sometimes even more expensive for patients with ultra-rare diseases, in part due to the limited number of patients. Pharmaceutical companies that develop a patented life-saving drug are in a position to charge a very high price, which, at best, may enable these companies to further develop drugs for use in rare disease. However, is there a limit to how much a life-saving drug should cost annually per patient? Government interventions and regulations may opt to withhold a life-saving drug solely due to its high price and cost-effectiveness. Processes related to drug pricing, reimbursement, and thereby availability, vary between countries, thus having implications on patient care. These processes are discussed, with specific focus on three drugs used in pediatric nephrology: agalsidase beta (for Fabry disease), eculizumab (for atypical hemolytic uremic syndrome), and cysteamine bitartrate (for cystinosis). Access to and costs of orphan drugs have most profound implications for patients, but also for their physicians, hospitals, insurance policies, and society at large, particularly from financial and ethical standpoints.
- Published
- 2016
- Full Text
- View/download PDF
33. Complement contributes to the pathogenesis of Shiga toxin–associated hemolytic uremic syndrome
- Author
-
Diana Karpman and Ramesh Tati
- Subjects
0301 basic medicine ,biology ,Lectin ,chemical and pharmacologic phenomena ,Shiga toxin ,bacterial infections and mycoses ,medicine.disease_cause ,Complement system ,Microbiology ,Pathogenesis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Renal injury ,Nephrology ,Immunology ,biology.protein ,Alternative complement pathway ,medicine ,Antibody ,Escherichia coli ,030215 immunology - Abstract
Complement is activated during Shiga toxin–producing Escherichia coli –associated hemolytic uremic syndrome (STEC-HUS). There is evidence of complement activation via the alternative pathway in STEC-HUS patients as well as from in vivo and in vitro models. Ozaki et al. demonstrate activation of the mannose-binding lectin (MBL) pathway in Shiga toxin–treated mice expressing human MBL2, but lacking murine Mbl s. Treatment with anti–human MBL2 antibody was protective, suggesting that MBL pathway activation also contributes to Shiga toxin–mediated renal injury.
- Published
- 2016
- Full Text
- View/download PDF
34. Shiga Toxin–Induced Complement-Mediated Hemolysis and Release of Complement-Coated Red Blood Cell–Derived Microvesicles in Hemolytic Uremic Syndrome
- Author
-
Anne-lie Ståhl, Ann-Charlotte Kristoffersson, Diana Karpman, Julia S. Westman, Minola Manea Hedström, Ida Arvidsson, Martin L. Olsson, Christian Rylander, and Jill R. Storry
- Subjects
Adult ,Male ,Erythrocytes ,Immunology ,Coated Vesicles ,Gene Expression ,Complement Membrane Attack Complex ,Suramin ,Biology ,Antibodies, Monoclonal, Humanized ,Escherichia coli O157 ,Hemolysis ,Complement factor B ,Shiga Toxin ,hemic and lymphatic diseases ,Purinergic P2 Receptor Antagonists ,medicine ,Humans ,Immunology and Allergy ,Child ,Complement Activation ,Edetic Acid ,Escherichia coli Infections ,Aged ,L-Lactate Dehydrogenase ,Receptors, Purinergic P2 ,Trihexosylceramides ,Infant ,Complement C3 ,Middle Aged ,Eculizumab ,Complement C9 ,medicine.disease ,Molecular biology ,Microvesicles ,Hemolytic Process ,Complement system ,Red blood cell ,medicine.anatomical_structure ,Biochemistry ,Child, Preschool ,Hemolytic-Uremic Syndrome ,Female ,Complement membrane attack complex ,medicine.drug - 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 P1k and P2k phenotype RBCs, expressing high levels of the Pk 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.
- Published
- 2015
- Full Text
- View/download PDF
35. Microvesicle Involvement in Shiga Toxin-Associated Infection
- Author
-
Diana Karpman, Ashmita Tontanahal, and Annie Villysson
- Subjects
0301 basic medicine ,Hemolytic anemia ,kidney ,Thrombotic microangiopathy ,Health, Toxicology and Mutagenesis ,lcsh:Medicine ,Review ,030204 cardiovascular system & hematology ,Biology ,Toxicology ,Virulence factor ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,enterohemorrhagic Escherichia coli ,Cell-Derived Microparticles ,medicine ,Animals ,Humans ,Escherichia coli Infections ,Kidney ,Microvesicle ,lcsh:R ,Shiga toxin ,medicine.disease ,Microvesicles ,Hemolysis ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Hemolytic-Uremic Syndrome ,biology.protein ,hemolytic uremic syndrome ,microvesicles - Abstract
Shiga toxin is the main virulence factor of enterohemorrhagic Escherichia coli, a non-invasive pathogen that releases virulence factors in the intestine, causing hemorrhagic colitis and, in severe cases, hemolytic uremic syndrome (HUS). HUS manifests with acute renal failure, hemolytic anemia and thrombocytopenia. Shiga toxin induces endothelial cell damage leading to platelet deposition in thrombi within the microvasculature and the development of thrombotic microangiopathy, mostly affecting the kidney. Red blood cells are destroyed in the occlusive capillary lesions. This review focuses on the importance of microvesicles shed from blood cells and their participation in the prothrombotic lesion, in hemolysis and in the transfer of toxin from the circulation into the kidney. Shiga toxin binds to blood cells and may undergo endocytosis and be released within microvesicles. Microvesicles normally contribute to intracellular communication and remove unwanted components from cells. Many microvesicles are prothrombotic as they are tissue factor- and phosphatidylserine-positive. Shiga toxin induces complement-mediated hemolysis and the release of complement-coated red blood cell-derived microvesicles. Toxin was demonstrated within blood cell-derived microvesicles that transported it to renal cells, where microvesicles were taken up and released their contents. Microvesicles are thereby involved in all cardinal aspects of Shiga toxin-associated HUS, thrombosis, hemolysis and renal failure.
- Published
- 2017
36. Extracellular vesicles in renal disease
- Author
-
Anne-lie Ståhl, Ida Arvidsson, and Diana Karpman
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Kidney ,End stage renal disease ,Nephropathy ,03 medical and health sciences ,Cystic kidney disease ,Extracellular Vesicles ,medicine ,Animals ,Humans ,Renal stem cell ,business.industry ,Acute kidney injury ,medicine.disease ,Microvesicles ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Nephrology ,Cancer research ,Kidney Diseases ,business ,Kidney disease - Abstract
Extracellular vesicles, such as exosomes and microvesicles, are host cell-derived packages of information that allow cell-cell communication and enable cells to rid themselves of unwanted substances. The release and uptake of extracellular vesicles has important physiological functions and may also contribute to the development and propagation of inflammatory, vascular, malignant, infectious and neurodegenerative diseases. This Review describes the different types of extracellular vesicles, how they are detected and the mechanisms by which they communicate with cells and transfer information. We also describe their physiological functions in cellular interactions, such as in thrombosis, immune modulation, cell proliferation, tissue regeneration and matrix modulation, with an emphasis on renal processes. We discuss how the detection of extracellular vesicles could be utilized as biomarkers of renal disease and how they might contribute to disease processes in the kidney, such as in acute kidney injury, chronic kidney disease, renal transplantation, thrombotic microangiopathies, vasculitides, IgA nephropathy, nephrotic syndrome, urinary tract infection, cystic kidney disease and tubulopathies. Finally, we consider how the release or uptake of extracellular vesicles can be blocked, as well as the associated benefits and risks, and how extracellular vesicles might be used to treat renal diseases by delivering therapeutics to specific cells.
- Published
- 2017
37. Aliskiren inhibits renin-mediated complement activation
- Author
-
Johan Rebetz, Ramesh Tati, Zivile Békássy, Ann-Charlotte Kristoffersson, Diana Karpman, and Anders I. Olin
- Subjects
0301 basic medicine ,Mast cell chemotaxis ,medicine.drug_class ,Glomerulonephritis, Membranoproliferative ,Complement C5b ,chemical and pharmacologic phenomena ,Complement C5a ,Cleavage (embryo) ,Renin inhibitor ,Complement factor B ,Opinions ,03 medical and health sciences ,chemistry.chemical_compound ,Fumarates ,Glomerular Basement Membrane ,Renin ,medicine ,Humans ,Mast Cells ,Child ,Complement Activation ,Kidney ,Chemotaxis ,Complement C4 ,Complement C3 ,Aliskiren ,Molecular biology ,Amides ,C3-convertase ,Complement system ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Nephrology ,Complement C3b ,Complement C3a ,Complement Factor D ,Female ,Complement Factor B - 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.
- Published
- 2017
38. C1-Inhibitor Decreases the Release of Vasculitis-Like Chemotactic Endothelial Microvesicles
- Author
-
Ramesh Tati, Ann-Charlotte Kristoffersson, L. M. Fredrik Leeb-Lundberg, Mårten Segelmark, Diana Karpman, Caroline Heijl, Anne-lie Ståhl, Maria Mossberg, and Robin Kahn
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.diagnostic_test ,medicine.drug_class ,Microvesicle ,Inflammation ,Chemotaxis ,General Medicine ,030204 cardiovascular system & hematology ,Kinin ,Biology ,Receptor antagonist ,Microvesicles ,Flow cytometry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Basic Research ,Nephrology ,Internal medicine ,medicine ,medicine.symptom ,Receptor - 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
- Published
- 2017
39. Atypical hemolytic uremic syndrome and C3 glomerulopathy: conclusions from a 'Kidney Disease: Improving Global Outcomes' (KDIGO) Controversies Conference
- Author
-
Richard J.H. Smith, Mireya Carratala, Mohamed R. Daha, Piero Ruggenenti, Gema Ariceta, Christoph Licht, Sanjeev Sethi, Mihály Józsi, Matthew C. Pickering, Charlie E. Alpers, Fadi Fakhouri, Gianluigi Ardissino, Marie-Agnès Dragon-Durey, Carla M. Nester, B. Paul Morgan, Nicole C. A. J. van de Kar, Jenna L.H. Smith, Fabrizio Spoleti, Marina Vivarelli, Linda Burke, Peter F. Zipfel, Lubka T. Roumenina, Marion Rabant, H. Terence Cook, Lynne D. Lanning, Moglie Le Quintrec, Michelle M. O’Shaughnessy, Fernando C. Fervenza, Eric Rondeau, David J. Kavanagh, Neil S. Sheerin, Hermann Haller, Mustafa Arici, Gerald B. Appel, Diana Karpman, Timothy H.J. Goodship, Agnes B. Fogo, Thomas D. Cairns, Marina Noris, Chantal Loirat, Arvind Bagga, Joshua M. Thurman, Sally Johnson, Santiago Rodríguez de Córdoba, Véronique Frémeaux-Bacchi, Daniel P. Gale, Ingeborg M. Bajema, An S. De Vriese, Vivette D. D'Agati, Francisco Monfort, Miguel Blasco, Laure Hélène Noël, Miriam Galbusera, Centre de Recherche en Transplantation et Immunologie (U1064 Inserm - CRTI), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Centre hospitalier universitaire de Nantes (CHU Nantes), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Complément et Maladies (CRC - Inserm U1138), Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Descartes - Paris 5 (UPD5), Université Pierre et Marie Curie - Paris 6 - UFR de Médecine Pierre et Marie Curie (UPMC), Université Pierre et Marie Curie - Paris 6 (UPMC), Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE), Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (APHP), Université Paris Diderot - Paris 7 (UPD7)-École pratique des hautes études (EPHE)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Diderot - Paris 7 (UPD7)-École pratique des hautes études (EPHE)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institute of Genetic Medicine [Newcastle, U.K.], Newcastle University [Newcastle], Centre for Complement and Inflammation Research [London, UK] (Department of Medicine), Imperial College London, Département de néphrologie et d'immunologie [CHU Nantes], Department of Nephrology and Hypertension [Rochester, MN, USA], Mayo Clinic [Rochester], Molecular Otolaryngology and Renal Research Laboratories [Iowa City, IA, USA] (Carver College of Medicine), University of Iowa [Iowa City]-Carver College of Medicine, University of Iowa, Department of Internal Medicine [Iowa City], Carver College of Medicine [Iowa City], University of Iowa [Iowa City]-University of Iowa [Iowa City], RCCS–Istituto di Ricerche Farmacologiche 'Mario Negri [Bergamo, Italy], Clinical Research Center for Rare Diseases 'Aldo e Cele Daccò' [Bergamo, Italy], Centro de Investigaciones Biológicas (CSIC), Consejo Superior de Investigaciones Científicas [Madrid] (CSIC), Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Université Sorbonne Paris Cité (USPC), Department of Laboratory Medicine and Pathology (Mayo Clinic Rochester), École pratique des hautes études (EPHE), and Le Bihan, Sylvie
- Subjects
IGA NEPHROPATHY ,Atypical hemolytic uremic syndrome ,kidney disease ,030232 urology & nephrology ,Disease ,030204 cardiovascular system & hematology ,[SDV.MHEP.UN]Life Sciences [q-bio]/Human health and pathology/Urology and Nephrology ,COMPLEMENT FACTOR-H ,Glomerulonephritis ,0302 clinical medicine ,C3 glomerulopathy ,complement ,0303 health sciences ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,RENAL-TRANSPLANTATION ,Treatment options ,Kidney disease ,Urology & Nephrology ,3. Good health ,Renal pathology ,Nephrology ,anti-complement therapies ,Life Sciences & Biomedicine ,medicine.medical_specialty ,OXFORD CLASSIFICATION ,Complement ,Disease pathogenesis ,Key issues ,ALTERNATIVE PATHWAY ,03 medical and health sciences ,Glomerulopathy ,medicine ,Intensive care medicine ,030304 developmental biology ,Anti-complement therapies ,Science & Technology ,business.industry ,atypical hemolytic uremic syndrome ,1103 Clinical Sciences ,medicine.disease ,MACULAR DEGENERATION ,Renal disorders Radboud Institute for Molecular Life Sciences [Radboudumc 11] ,MEMBRANOPROLIFERATIVE GLOMERULONEPHRITIS ,Immunology ,DENSE-DEPOSIT DISEASE ,business ,ECULIZUMAB MAINTENANCE TREATMENT ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,glomerulonephritis ,MESANGIOCAPILLARY GLOMERULONEPHRITIS - Abstract
13 p.-3 fig. Goodship, Timothy H.J. et al., In both atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy (C3G) complement plays a primary role in disease pathogenesis. Herein we report the outcome of a 2015 Kidney Disease: Improving Global Outcomes (KDIGO)Controversies Conference where key issues in the management of these 2 diseases were considered by a global panel of experts. Areas addressed included renal pathology, clinical phenotype and assessment, genetic drivers of disease, acquired drivers of disease, and treatment strategies. In order to help guide clinicians who are caring for such patients, recommendations for best treatment strategies were discussed at length, providing the evidence base underpinning current treatment options. Knowledge gaps were identified and a prioritized research agenda was proposed to resolve outstanding controversial issues., The conference was sponsored by Kidney Disease: Improving Global Outcomes (KDIGO) and supported in part by unrestricted educational grants from Achillion Pharmaceuticals, Akari Therapeutics, Alexion Pharmaceuticals, and Omeros.
- Published
- 2017
- Full Text
- View/download PDF
40. Ouabain Protects against Shiga Toxin–Triggered Apoptosis by Reversing the Imbalance between Bax and Bcl-xL
- Author
-
Hjalmar Brismar, Diana Karpman, Ievgeniia Burlaka, Anita Aperia, Liping Yang, Ida Arvidsson, Johan Rebetz, and Xiao Li Liu
- Subjects
Male ,bcl-X Protein ,Apoptosis ,Bcl-xL ,Caspase 3 ,In Vitro Techniques ,urologic and male genital diseases ,Caspase 8 ,Shiga Toxin 2 ,Ouabain ,Kidney Tubules, Proximal ,Rats, Sprague-Dawley ,Mice ,Bcl-2-associated X protein ,Downregulation and upregulation ,medicine ,Animals ,Inositol 1,4,5-Trisphosphate Receptors ,Enzyme Inhibitors ,Cells, Cultured ,bcl-2-Associated X Protein ,Kidney ,Dose-Response Relationship, Drug ,biology ,NF-kappa B ,General Medicine ,Molecular biology ,Rats ,Mice, Inbred C57BL ,Disease Models, Animal ,Basic Research ,medicine.anatomical_structure ,Nephrology ,biology.protein ,Sodium-Potassium-Exchanging ATPase ,Signal Transduction ,medicine.drug - Abstract
Hemolytic uremic syndrome, a life-threatening disease often accompanied by acute renal failure, usually occurs after gastrointestinal infection with Shiga toxin 2 (Stx2)-producing Escherichia coli. Stx2 binds to the glycosphingolipid globotriaosylceramide receptor, expressed by renal epithelial cells, and triggers apoptosis by activating the apoptotic factor Bax. Signaling via the ouabain/Na,K-ATPase/IP3R/NF-κB pathway increases expression of Bcl-xL, an inhibitor of Bax, suggesting that ouabain might protect renal cells from Stx2-triggered apoptosis. Here, exposing rat proximal tubular cells to Stx2 in vitro resulted in massive apoptosis, upregulation of the apoptotic factor Bax, increased cleaved caspase-3, and downregulation of the survival factor Bcl-xL; co-incubation with ouabain prevented all of these effects. Ouabain activated the NF-κB antiapoptotic subunit p65, and the inhibition of p65 DNA binding abolished the antiapoptotic effect of ouabain in Stx2-exposed tubular cells. Furthermore, in vivo, administration of ouabain reversed the imbalance between Bax and Bcl-xL in Stx2-treated mice. Taken together, these results suggest that ouabain can protect the kidney from the apoptotic effects of Stx2.
- Published
- 2013
- Full Text
- View/download PDF
41. An international consensus approach to the management of atypical hemolytic uremic syndrome in children
- Author
-
Daniel Landau, Magdalena Riedl, Chantal Loirat, Marina Vivarelli, Anne Laure Lapeyraque, Rosanna Coppo, Martin Bitzan, Sally Johnson, Anna Bjerre, Craig B. Langman, Francesco Emma, Véronique Frémeaux-Bacchi, Diana Karpman, Christoph Licht, Carmine Pecoraro, Gema Ariceta, Nicole C. A. J. van de Kar, Nesrin Besbas, Johan Vande Walle, Carla M. Nester, Fadi Fakhouri, Service de Néphrologie pédiatrique [Hôpital Robert Debré, Paris], Hôpital Robert Debré-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Centre de Recherche en Transplantation et Immunologie (U1064 Inserm - CRTI), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Département de néphrologie et d'immunologie [CHU Nantes], Centre hospitalier universitaire de Nantes (CHU Nantes), Institut de transplantation urologie-néphrologie (ITUN), Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes), Pediatric Nephrology [Barcelona, Spain] (Vall d’Hebron Hospital), Universitat Autònoma de Barcelona (UAB)-Vall d'Hebron University Hospital [Barcelona], Department of Pediatric Nephrology [Ankara, Turkey] ( Faculty of Medicine), Hacettepe University = Hacettepe Üniversitesi, Division of Nephrology [Montreal, Canada] (Montreal Children’s Hospital), Montreal Children's Hospital, McGill University Health Center [Montreal] (MUHC)-McGill University Health Center [Montreal] (MUHC), Department of Pediatrics [Oslo, Norway], Oslo University Hospital [Oslo], Science of Turin Health Agency [Turin, Italy] (City of the Health), Regina Margherita University Children's Hospital [Turin, Italy], Bambino Gesù Children’s Hospital [Rome, Italy], Department of Paediatric Nephrology [Newcastle Upon Tyne, UK], Great North Children’s Hospital [Newcastle Upon Tyne, UK], Department of Pediatrics [Lund, Sweden] (Clinical Sciences), Lund University [Lund], Soroka University Medical Center [Beer Sheva, Israel], Ann & Robert H. Lurie Children's Hospital of Chicago, Service de Néphrologie [Montréal, Canada] (Département de Pédiatrie), Université de Montréal (UdeM), The Hospital for sick children [Toronto] (SickKids), the Stead Family Department of Pediatrics [Iowa City, Iowa, USA] (Departments of Internal Medicine), University of Iowa [Iowa City], Santobono Children’s Hospital [Naples, Italy], Department of Pediatrics [Innsbruck, Austria], Innsbruck Medical University [Austria] (IMU), Department of Pediatric Nephrology [Nijmegen, The Netherlands] ( Amalia Children’s Hospital), Radboud University Medical Center [Nijmegen]-Amalia Children’s Hospital [Nijmegen, The Netherlands], Ghent University Hospital [Ghent, Belgium], Immunologie et Cancérologie Intégratives (CRC - Inserm U1138), Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Le Bihan, Sylvie, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Robert Debré, and Innsbruck Medical University = Medizinische Universität Innsbruck (IMU)
- Subjects
Anti-factor H antibody ,Nephrology ,Atypical hemolytic uremic syndrome ,International Cooperation ,030232 urology & nephrology ,Disease ,030204 cardiovascular system & hematology ,Kidney transplantation ,0302 clinical medicine ,Risk Factors ,Hemolytic uremic syndrome ,Cooperative Behavior ,Child ,Complement Activation ,Children ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Age Factors ,Eculizumab ,Combined Modality Therapy ,3. Good health ,Treatment Outcome ,Child, Preschool ,Combined liver–kidney transplantation ,Drug Monitoring ,medicine.drug ,medicine.medical_specialty ,Consensus ,Thrombotic microangiopathy ,Adolescent ,Complement ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,Therapeutic approach ,Monitoring, Immunologic ,Predictive Value of Tests ,Internal medicine ,medicine ,Humans ,Immunologic Factors ,Plasma exchange ,Intensive care medicine ,business.industry ,Patient Selection ,Infant, Newborn ,Infant ,medicine.disease ,Liver Transplantation ,Transplantation ,Renal disorders Radboud Institute for Molecular Life Sciences [Radboudumc 11] ,Pediatrics, Perinatology and Child Health ,Plasma infusion ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Item does not contain fulltext Atypical hemolytic uremic syndrome (aHUS) emerged during the last decade as a disease largely of complement dysregulation. This advance facilitated the development of novel, rational treatment options targeting terminal complement activation, e.g., using an anti-C5 antibody (eculizumab). We review treatment and patient management issues related to this therapeutic approach. We present consensus clinical practice recommendations generated by HUS International, an international expert group of clinicians and basic scientists with a focused interest in HUS. We aim to address the following questions of high relevance to daily clinical practice: Which complement investigations should be done and when? What is the importance of anti-factor H antibody detection? Who should be treated with eculizumab? Is plasma exchange therapy still needed? When should eculizumab therapy be initiated? How and when should complement blockade be monitored? Can the approved treatment schedule be modified? What approach should be taken to kidney and/or combined liver-kidney transplantation? How should we limit the risk of meningococcal infection under complement blockade therapy? A pressing question today regards the treatment duration. We discuss the need for prospective studies to establish evidence-based criteria for the continuation or cessation of anticomplement therapy in patients with and without identified complement mutations.
- Published
- 2016
- Full Text
- View/download PDF
42. Editorial Commentary: Escherichia coli O104:H4 and Hemolytic Uremic Syndrome: The Analysis Begins
- Author
-
Diana Karpman and Phillip I. Tarr
- Subjects
Male ,Microbiology (medical) ,Shiga-Toxigenic Escherichia coli ,business.industry ,Disease Outbreaks ,Microbiology ,Infectious Diseases ,Escherichia coli O104:H4 ,Hemolytic-Uremic Syndrome ,Humans ,Medicine ,Female ,business - Published
- 2012
- Full Text
- View/download PDF
43. Cross-Reactive Protection against Enterohemorrhagic Escherichia coli Infection by Enteropathogenic E. coli in a Mouse Model
- Author
-
Ida Arvidsson, Diana Karpman, and Carla Calderon Toledo
- Subjects
Cross Protection ,Immunoblotting ,Immunology ,Biology ,medicine.disease_cause ,digestive system ,Pediatrics ,Microbiology ,Enteropathogenic Escherichia coli ,Mice ,Immunity ,parasitic diseases ,medicine ,Animals ,Escherichia coli ,Escherichia coli Infections ,Intimin ,Bacterial Shedding ,Mice, Inbred BALB C ,Body Weight ,Bacterial Infections ,biochemical phenomena, metabolism, and nutrition ,bacterial infections and mycoses ,biology.organism_classification ,Antibodies, Bacterial ,Pathogenicity island ,Enterobacteriaceae ,Virology ,Infectious Diseases ,Enterohemorrhagic Escherichia coli ,biology.protein ,bacteria ,Parasitology ,Antibody ,Locus of enterocyte effacement - Abstract
Enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) are related attaching and effacing (A/E) pathogens. The genes responsible for the A/E pathology are carried on a chromosomal pathogenicity island termed the locus of enterocyte effacement (LEE). Both pathogens share a high degree of homology in the LEE and additional O islands. EHEC prevalence is much lower in areas where EPEC is endemic. This may be due to the development of antibodies against common EPEC and EHEC antigens. This study investigated the hypothesis that EPEC infections may protect against EHEC infections. We used a mouse model to inoculate BALB/c mice intragastrically, first with EPEC and then with EHEC ( E. coli O157:H7). Four control groups received either a nonpathogenic E. coli (NPEC) strain followed by EHEC (NPEC/EHEC), phosphate-buffered saline (PBS) followed by EHEC (PBS/EHEC), EPEC/PBS, or PBS/PBS. Mice were monitored for weight loss and symptoms. EPEC colonized the intestine after challenge, and mice developed serum antibodies to intimin and E. coli secreted protein B (encoded in the LEE). Prechallenge with an EPEC strain had a protective effect after EHEC infection, as only a few mice developed mild symptoms, from which they recovered. These mice had an increase in body weight similar to that in control animals, and tissue morphology exhibited mild intestinal changes and normal renal histology. All mice that were not prechallenged with the EPEC strain developed mild to severe symptoms after EHEC infection, with weight loss as well as intestinal and renal histopathological changes. These data suggest that EPEC may protect against EHEC infection in this mouse model.
- Published
- 2011
- Full Text
- View/download PDF
44. Complement activation on platelet-leukocyte complexes and microparticles in enterohemorrhagic Escherichia coli–induced hemolytic uremic syndrome
- Author
-
Anne-lie Ståhl, Diana Karpman, and Lisa Sartz
- Subjects
Blood Platelets ,Lipopolysaccharides ,Male ,Lipopolysaccharide ,Neutrophils ,Complement Pathway, Alternative ,Immunology ,Population ,Complement Membrane Attack Complex ,In Vitro Techniques ,Biology ,Escherichia coli O157 ,Shiga Toxin 1 ,Shiga Toxin 2 ,Biochemistry ,Monocytes ,Microbiology ,chemistry.chemical_compound ,Phagocytosis ,Cell-Derived Microparticles ,medicine ,Humans ,Platelet ,Platelet activation ,Child ,education ,Complement Activation ,Escherichia coli Infections ,Whole blood ,education.field_of_study ,Monocyte ,Infant ,Shiga toxin ,Complement C3 ,Cell Biology ,Hematology ,Complement C9 ,Complement system ,medicine.anatomical_structure ,chemistry ,Child, Preschool ,Hemolytic-Uremic Syndrome ,Complement C3a ,biology.protein ,Female - Abstract
Hemolytic uremic syndrome (HUS) is commonly associated with Shiga toxin (Stx)–producing Escherichia coli O157:H7 infection. This study examined patient samples for complement activation on leukocyte-platelet complexes and microparticles, as well as donor samples for Stx and lipopolysaccharide (O157LPS)–induced complement activation on platelet-leukocyte complexes and microparticles. Results, analyzed by flow cytometry, showed that whole blood from a child with HUS had surface-bound C3 on 30% of platelet-monocyte complexes compared with 14% after recovery and 12% in pediatric controls. Plasma samples from 12 HUS patients were analyzed for the presence of microparticles derived from platelets, monocytes, and neutrophils. Acute-phase samples exhibited high levels of platelet microparticles and, to a lesser extent, monocyte microparticles, both bearing C3 and C9. Levels decreased significantly at recovery. Stx or O157LPS incubated with donor whole blood increased the population of platelet-monocyte and platelet-neutrophil complexes with surface-bound C3 and C9, an effect enhanced by costimulation with Stx and O157LPS. Both Stx and O157LPS induced the release of C3- and C9-bearing microparticles from platelets and monocytes. Released microparticles were phagocytosed by neutrophils. The presence of complement on platelet-leukocyte complexes and microparticles derived from these cells suggests a role in the inflammatory and thrombogenic events that occur during HUS.
- Published
- 2011
- Full Text
- View/download PDF
45. Kinin system activation in vasculitis*
- Author
-
Robin Kahn and Diana Karpman
- Subjects
business.industry ,Bradykinin ,Inflammation ,General Medicine ,Kinin ,medicine.disease ,chemistry.chemical_compound ,chemistry ,Proteinase 3 ,Vasoactive ,Pediatrics, Perinatology and Child Health ,Tissue damage ,Immunology ,cardiovascular system ,Autoimmune inflammatory disease ,Medicine ,cardiovascular diseases ,medicine.symptom ,business ,Vasculitis ,circulatory and respiratory physiology - Abstract
Vasculitis is a systemic autoimmune inflammatory disease, characterized by inflammation in and around vessel walls leading to perturbed vessel patency and tissue damage. Many different organs may be involved. In this review pathogenetic mechanisms of vasculitis are discussed, with special reference to activation of the kinin system. Mechanisms of kinin system activation are described ultimately leading to release of kinins from high-molecular-weight-kininogen. These vasoactive peptides promote inflammation. Conclusion: Kinin system activation during vasculitis promotes inflammation.
- Published
- 2011
- Full Text
- View/download PDF
46. Hyperfiltration evaluated by glomerular filtration rate at diagnosis in children with cancer
- Author
-
Thomas Wiebe, Diana Karpman, and Lars Hjorth
- Subjects
Chemotherapy ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Urology ,Cancer ,Kidney metabolism ,Renal function ,Hematology ,Urine ,urologic and male genital diseases ,medicine.disease ,Pediatric cancer ,Group B ,Endocrinology ,Oncology ,Internal medicine ,Pediatrics, Perinatology and Child Health ,medicine ,business ,Prospective cohort study - Abstract
BACKGROUND: Renal glomerular filtration rate (GFR) of pediatric cancer patients at diagnosis has previously been investigated in a limited number of studies. PROCEDURE: GFR, measured by iohexol clearance, was prospectively investigated in 55 children over the age of 1 year with malignancies, (group A). Elevated GFR (>175 ml/min/1.73 m(2)) at diagnosis was found. To investigate if this finding was consistent, a second group of 76 children with malignancies was studied, (group B). As a method control for GFR obtained by iohexol clearance, group A and B together were compared to 298 pediatric patients without cancer, group C. RESULTS: GFR was elevated in 40/131 (31%) in Group A + B but only in 17/298 (6%) in Group C. GFR was significantly higher in children aged 1-5 in group A + B (47%) compared to group C (3%). Bone marrow involvement was significantly associated with higher GFR. Children without bone marrow involvement also hyperfiltrated more often than controls, but less often. Urea in urine was used as a marker of renal protein clearance in 14 patients in group A. A significant correlation between u-urea (mmol/L)/u-creatinine (mmol/L) and GFR was noted. CONCLUSIONS: Hyperfiltration is sometimes present in children with cancer at diagnosis. This may be related to increased amino acid turn over in patients with a large tumor burden. An elevated initial GFR in a child with cancer, which normalizes after chemotherapy may indicate chemotherapy-induced decreased renal function, but can be due to normalization of an initially high GFR. Pediatr Blood Cancer © 2011 Wiley-Liss, Inc. (Less)
- Published
- 2011
- Full Text
- View/download PDF
47. IgA nephropathy associated with a novel N-terminal mutation in factor H
- Author
-
Ann-Charlotte Kristoffersson, Rafael T. Krmar, Diana Karpman, Magnus Söderberg, and Roland Schmitt
- Subjects
medicine.medical_specialty ,Adolescent ,Complement factor I ,medicine.disease_cause ,Immunofluorescence ,Pediatrics ,Nephropathy ,Diagnosis, Differential ,Hypertension, Malignant ,Internal medicine ,medicine ,Humans ,Mutation ,Proteinuria ,Purpura, Thrombotic Thrombocytopenic ,medicine.diagnostic_test ,business.industry ,Glomerulonephritis, IGA ,Glomerulonephritis ,Microangiopathic hemolytic anemia ,medicine.disease ,Endocrinology ,Complement Factor H ,Pediatrics, Perinatology and Child Health ,Female ,Renal biopsy ,medicine.symptom ,business - Abstract
Most patients with IgA nephropathy exhibit complement deposition in the glomerular mesangium. Certain cases of IgA nephropathy have been associated with reduced levels of complement factor H. A recent study could not demonstrate mutations at the C-terminal of factor H. We describe a novel heterozygous mutation in factor H, position A48S (nucleotide position 142 G > T, alanine > serine), detected in exon 2 of a 14-year-old girl with IgA nephropathy. The patient exhibited reduced levels of C3 and factor H, the latter suggesting that the mutation affected factor H secretion. The patient developed initial signs and symptoms of glomerulonephritis at the age of 9 years but presented again at the age of 14 years with weight gain, renal failure, nephrotic-range proteinuria and malignant hypertension. Blood tests suggested the development of microangiopathic hemolytic anemia (MAHA) but the renal biopsy was mostly indicative of chronic changes associated with IgA nephropathy as well as vascular changes associated with malignant hypertension. Immunofluorescence exhibited deposits of IgA, C3, and IgM. Screening of the factor H gene revealed, in addition to the mutation, three heterozygous hemolytic uremic syndrome -associated risk polymorphisms (-257 c/t, 2089 a/g, and 2881 g/t) which may have increased the patient's susceptibility to the occurrence of MAHA triggered by malignant hypertension. The combined clinical picture of IgA nephropathy and MAHA may have been partly related to the alterations in factor H.
- Published
- 2010
- Full Text
- View/download PDF
48. A novel mutation in the complement regulator clusterin in recurrent hemolytic uremic syndrome
- Author
-
Anders I. Olin, Anne-lie Ståhl, Willem Proesmans, Anne-Marie Roodhooft, Ann-Charlotte Kristoffersson, Martin L. Olsson, and Diana Karpman
- Subjects
Blood Platelets ,Male ,Adolescent ,CD40 Ligand ,Immunology ,Mutant ,Biology ,medicine.disease_cause ,Hemolysis ,Exon ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Platelet activation ,Child ,Molecular Biology ,Mutation ,Clusterin ,CD46 ,Complement C5 ,Complement C3 ,Complement System Proteins ,Exons ,Complement C9 ,Molecular biology ,Complement system ,Child, Preschool ,Immunoglobulin G ,COS Cells ,Hemolytic-Uremic Syndrome ,biology.protein ,Female ,Rabbits ,Complement membrane attack complex ,Protein Binding - Abstract
A novel heterozygous mutation in the clusterin gene, nucleotide position A1298C (glutamine > proline Q433P), was detected in exon 7 of a child with recurrent hemolytic uremic syndrome (HUS). The same mutation was found in the child's two siblings and mother but not in 120 controls. In addition, a previously described heterozygous mutation was detected in the gene encoding membrane cofactor protein (MCP) causing a 6 base-pair deletion 811-816delGACAGT in exon 6. It was found in the patient, both siblings and the father. One sibling had recovered from post-streptoccocal glomerulonephritis. Clusterin levels in the patient, siblings and parents were normal as was the migration pattern in a gel. Patient serum induced C3 and C9 deposition on normal washed platelets, and platelet activation, as detected by flow cytometry. The same phenomenon was found in serum taken from the siblings and the mother but not in the sample from the father and controls. Addition of clusterin to patient serum did not inhibit complement activation on platelets. The Q433P mutant, in isolated form, was further studied by binding to the components of the terminal complement complex. The mutant did not bind to C5b-7 that was immobilized onto a BIAcore chip, whereas wild-type clusterin did, indicating that the mutation could lead to defective inhibition of formation of the membrane attack complex under these conditions. Hemolysis of rabbit erythrocytes was inhibited by wild-type clusterin but not by the mutant. Mutated clusterin could thus not prevent assembly of the membrane attack complex on platelets and erythrocytes.
- Published
- 2009
- Full Text
- View/download PDF
49. Neutrophil-Derived Proteinase 3 Induces Kallikrein-Independent Release of a Novel Vasoactive Kinin
- Author
-
Mihail Todiras, Thomas Hellmark, Diana Karpman, Kerstin Westman, Robin Kahn, L. M. Fredrik Leeb-Lundberg, Anders Christensson, Nasrin Akbari, Jörgen Wieslander, Tor Olofsson, Werner Müller-Esterl, and Michael Bader
- Subjects
Male ,medicine.medical_specialty ,Adolescent ,Neutrophils ,Myeloblastin ,Proteolysis ,Immunology ,Bradykinin ,Inflammation ,Kinins ,Biology ,Mice ,chemistry.chemical_compound ,Proteinase 3 ,Internal medicine ,medicine ,Animals ,Humans ,Immunology and Allergy ,cardiovascular diseases ,Child ,Receptor ,Aged ,Aged, 80 and over ,Kininogen ,medicine.diagnostic_test ,Kininogens ,Kallikrein ,Middle Aged ,Kinin ,Molecular biology ,Rats ,Endocrinology ,chemistry ,Kallikreins ,medicine.symptom ,Protein Binding ,circulatory and respiratory physiology - Abstract
The kinin-forming pathway is activated on endothelial cells and neutrophils when high-molecular weight kininogen (HK) is cleaved by plasma kallikrein liberating bradykinin, a potent mediator of inflammation. Kinins are released during inflammatory conditions such as vasculitis, associated with neutrophil influx around blood vessels. Some patients with vasculitis have elevated plasma levels of neutrophil-derived proteinase 3 (PR3) and anti-PR3 Abs. This study investigated if neutrophil-derived PR3 could induce activation of the kinin pathway. PR3 incubated with HK, or a synthetic peptide derived from HK, induced breakdown and release of a novel tridecapeptide termed PR3-kinin, NH2-MKRPPGFSPFRSS-COOH, consisting of bradykinin with two additional amino acids on each terminus. The reaction was specific and inhibited by anti-PR3 and α1-antitrypsin. Recombinant wild-type PR3 incubated with HK induced HK breakdown, whereas mutated PR3, lacking enzymatic activity, did not. PR3-kinin bound to and activated human kinin B1 receptors, but did not bind to B2 receptors, expressed by transfected HEK293 cells in vitro. In human plasma PR3-kinin was further processed to the B2 receptor agonist bradykinin. PR3-kinin exerted a hypotensive effect in vivo through both B1 and B2 receptors as demonstrated using wild-type and B1 overexpressing rats as well as wild-type and B2 receptor knockout mice. Neutrophil extracts from vasculitis patients and healthy controls contained comparable amounts of PR3 and induced HK proteolysis, an effect that was abolished when PR3 was immunoadsorbed. Neutrophil-derived PR3 can proteolyze HK and liberate PR3-kinin, thereby initiating kallikrein-independent activation of the kinin pathway.
- Published
- 2009
- Full Text
- View/download PDF
50. Molecular basis of ADAMTS13 dysfunction in thrombotic thrombocytopenic purpura
- Author
-
Diana Karpman and Minola Manea
- Subjects
Hemolytic anemia ,medicine.medical_specialty ,Endothelium ,Thrombotic thrombocytopenic purpura ,ADAMTS13 Protein ,Diagnosis, Differential ,Von Willebrand factor ,hemic and lymphatic diseases ,Internal medicine ,von Willebrand Factor ,medicine ,Humans ,Platelet ,Kidney ,Purpura, Thrombotic Thrombocytopenic ,biology ,business.industry ,medicine.disease ,ADAMTS13 ,Schistocyte ,ADAM Proteins ,medicine.anatomical_structure ,Endocrinology ,Nephrology ,Mutation ,Pediatrics, Perinatology and Child Health ,Immunology ,cardiovascular system ,biology.protein ,business ,circulatory and respiratory physiology - Abstract
Thrombotic thrombocytopenic purpura (TTP) is a thrombotic microangiopathic disorder characterized by thrombocytopenia, hemolytic anemia, neurological and renal manifestations, and fever. It is associated with dysfunctional von Willebrand factor (VWF) proteolysis and the occurrence of VWF- and platelet-rich thrombi in the microcirculation of multiple organs, including the kidneys. Von Willebrand factor is a large glycoprotein that circulates in plasma as a series of multimers, and it plays a major role in primary hemostasis by inducing the formation of platelet plugs at sites of vascular injury and high-shear stress. Its activity is dependent on the sizes of the multimers, with ultra-large (UL) VWF multimers being biologically very potent. The ULVWF multimers are rapidly degraded upon their secretion from endothelial cells in normal individuals but not in the circulation of TTP patients, causing the formation of disseminated thrombi in the latter. The defective breakdown of VWF is attributed to a severely deficient activity of the VWF-cleaving protease ADAMTS13, a plasma metalloprotease synthesized in the liver, kidneys, and endothelium. This protease rapidly degrades VWF-platelet strings under flow by proteolytic cleavage of the VWF subunit, thereby regulating the size of the platelet thrombus. Congenital TTP occurs due to ADAMTS13 mutations, with the usual debut occurring during the first years of life, while acquired TTP is associated with auto-antibodies against ADAMTS13.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.