Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article En | MEDLINE | ID: mdl-38673910

Endothelial cell (EC) injury is a crucial contributor to the progression of diabetic kidney disease (DKD), but the specific EC populations and mechanisms involved remain elusive. Kidney ECs (n = 5464) were collected at three timepoints from diabetic BTBRob/ob mice and non-diabetic littermates. Their heterogeneity, transcriptional changes, and alternative splicing during DKD progression were mapped using SmartSeq2 single-cell RNA sequencing (scRNAseq) and elucidated through pathway, network, and gene ontology enrichment analyses. We identified 13 distinct transcriptional EC phenotypes corresponding to different kidney vessel subtypes, confirmed through in situ hybridization and immunofluorescence. EC subtypes along nephrons displayed extensive zonation related to their functions. Differential gene expression analyses in peritubular and glomerular ECs in DKD underlined the regulation of DKD-relevant pathways including EIF2 signaling, oxidative phosphorylation, and IGF1 signaling. Importantly, this revealed the differential alteration of these pathways between the two EC subtypes and changes during disease progression. Furthermore, glomerular and peritubular ECs also displayed aberrant and dynamic alterations in alternative splicing (AS), which is strongly associated with DNA repair. Strikingly, genes displaying differential transcription or alternative splicing participate in divergent biological processes. Our study reveals the spatiotemporal regulation of gene transcription and AS linked to DKD progression, providing insight into pathomechanisms and clues to novel therapeutic targets for DKD treatment.


Alternative Splicing , Diabetic Nephropathies , Endothelial Cells , Single-Cell Analysis , Transcriptome , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Mice , Single-Cell Analysis/methods , Endothelial Cells/metabolism , Endothelial Cells/pathology , Kidney/metabolism , Kidney/pathology , Gene Expression Regulation , Transcription, Genetic , Gene Expression Profiling/methods , Male
3.
Int J Mol Sci ; 23(23)2022 Dec 01.
Article En | MEDLINE | ID: mdl-36499397

Deciphering the pathophysiological mechanisms of primary podocytopathies that can lead to end-stage renal disease and increased mortality is an unmet need. Studying how microRNAs (miRs) interfere with various signaling pathways enables identification of pathomechanisms, novel biomarkers and potential therapeutic options. We investigated the expression of miR-200c in urine from patients with different renal diseases as a potential candidate involved in podocytopathies. The role of miR-200c for the glomerulus and its potential targets were studied in cultured human podocytes, human glomerular endothelial cells and in the zebrafish model. miR-200c was upregulated in urine from patients with minimal change disease, membranous glomerulonephritis and focal segmental glomerulosclerosis and also in transforming growth factor beta (TGF-ß) stressed glomerular endothelial cells, but not in podocytes. In zebrafish, miR-200c overexpression caused proteinuria, edema, podocyte foot process effacement and glomerular endotheliosis. Although zinc finger E-Box binding homeobox 1/2 (ZEB1/2), important in epithelial to mesenchymal transition (EMT), are prominent targets of miR-200c, their downregulation did not explain our zebrafish phenotype. We detected decreased vegfaa/bb in zebrafish overexpressing miR-200c and could further prove that miR-200c decreased VEGF-A expression and secretion in cultured human podocytes. We hypothesize that miR-200c is released from glomerular endothelial cells during cell stress and acts in a paracrine, autocrine, as well as context-dependent manner in the glomerulus. MiR-200c can cause glomerular damage most likely due to the reduction of podocyte VEGF-A. In contrast, miR-200c might also influence ZEB expression and therefore EMT, which might be important in other conditions. Therefore, we propose that miR-200c-mediated effects in the glomerulus are context-sensitive.


Endothelial Cells , MicroRNAs , Animals , Humans , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition , MicroRNAs/genetics , MicroRNAs/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/genetics , Zebrafish/metabolism
4.
EBioMedicine ; 47: 319-328, 2019 Sep.
Article En | MEDLINE | ID: mdl-31444145

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.


Complement Activation/immunology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Kallikrein-Kinin System/drug effects , Vasculitis/etiology , Vasculitis/metabolism , Adult , Aged , Animals , Biological Transport , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Cell-Derived Microparticles/metabolism , Cells, Cultured , Complement C1 Inhibitor Protein/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Models, Animal , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Immunoglobulin G/immunology , Male , Mice , Middle Aged , Protein Binding , Vasculitis/pathology
5.
Front Immunol ; 9: 1988, 2018.
Article En | MEDLINE | ID: mdl-30233584

Non-typeable Haemophilus influenzae (NTHi) is a Gram-negative human commensal commonly residing in the nasopharynx of preschool children. It occasionally causes upper respiratory tract infection such as acute otitis media, but can also spread to the lower respiratory tract causing bronchitis and pneumonia. There is increasing recognition that NTHi has an important role in chronic lower respiratory tract inflammation, particularly in persistent infection in patients suffering from chronic obstructive pulmonary disease (COPD). Here, we set out to assess the innate protective effects of collagen VI, a ubiquitous extracellular matrix component, against NTHi infection in vivo. In vitro, collagen VI rapidly kills bacteria through pore formation and membrane rupture, followed by exudation of intracellular content. This effect is mediated by specific binding of the von Willebrand A (VWA) domains of collagen VI to the NTHi surface adhesins protein E (PE) and Haemophilus autotransporter protein (Hap). Similar observations were made in vivo specimens from murine airways and COPD patient biopsies. NTHi bacteria adhered to collagen fibrils in the airway mucosa and were rapidly killed by membrane destabilization. The significance in host-pathogen interplay of one of these molecules, PE, was highlighted by the observation that it confers partial protection from bacterial killing. Bacteria lacking PE were more prone to antimicrobial activity than NTHi expressing PE. Altogether the data shed new light on the carefully orchestrated molecular events of the host-pathogen interplay in COPD and emphasize the importance of the extracellular matrix as a novel branch of innate host defense.


Collagen Type IV/metabolism , Extracellular Matrix/metabolism , Fibroblasts/physiology , Haemophilus Infections/immunology , Haemophilus influenzae/physiology , Lung/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Adhesins, Bacterial/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Female , Haemophilus Infections/complications , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Mice , Mice, Inbred C57BL , Protein Binding , Pulmonary Disease, Chronic Obstructive/complications
6.
Kidney Int ; 94(4): 689-700, 2018 10.
Article En | MEDLINE | ID: mdl-29884545

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.


Amides/pharmacology , Complement Activation/drug effects , Complement C3/chemistry , Fumarates/pharmacology , Glomerulonephritis, Membranoproliferative/metabolism , Glomerulonephritis, Membranoproliferative/therapy , Renin/chemistry , Amides/therapeutic use , Chemotaxis/drug effects , Child , Complement C3/metabolism , Complement C3a/chemistry , Complement C3a/metabolism , Complement C3b/chemistry , Complement C3b/metabolism , Complement C4/chemistry , Complement C5a/chemistry , Complement C5a/metabolism , Complement C5b/chemistry , Complement C5b/metabolism , Complement Factor B/chemistry , Complement Factor D/chemistry , Female , Fumarates/therapeutic use , Glomerular Basement Membrane/pathology , Glomerulonephritis, Membranoproliferative/pathology , Humans , Mast Cells/physiology , Renin/antagonists & inhibitors , Renin/metabolism
7.
J Immunol ; 201(3): 1007-1020, 2018 08 01.
Article En | MEDLINE | ID: mdl-29925677

Collagen VI is a ubiquitous extracellular matrix component that forms extensive microfibrillar networks in most connective tissues. In this study, we describe for the first time, to our knowledge, that the collagen VI von Willebrand factor type A-like domains exhibit a broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria in human skin infections in vivo. In silico sequence and structural analysis of VWA domains revealed that they contain cationic and amphipathic peptide sequence motifs, which might explain the antimicrobial nature of collagen VI. In vitro and in vivo studies show that these peptides exhibited significant antibacterial activity against Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa through membrane disruption. Our findings shed new light on the role of collagen VI-derived peptides in innate host defense and provide templates for development of peptide-based antibacterial therapies.


Anti-Bacterial Agents/immunology , Collagen Type VI/immunology , Peptides/immunology , Bacteria/immunology , Bacterial Infections/immunology , Humans , Immunity, Innate/immunology , Protein Domains/immunology , Skin/immunology , Skin/microbiology , Skin Diseases, Bacterial/immunology
8.
J Wound Care ; 27(4): 199-209, 2018 04 02.
Article En | MEDLINE | ID: mdl-29637827

OBJECTIVE: When the delicate balance between catabolic and anabolic processes is disturbed for any reason, the healing process can stall, resulting in chronic wounds. In chronic wound pathophysiology, proteolytic imbalance is implicated due to elevated protease levels mediating tissue damage. Hence, it is important to design appropriate wound treatments able to control and modulate protease activity directly at the host/biomaterial interface. Here, we investigate collagen-based wound dressings with the focus on their potential to adsorb and inactivate tissue proteases. METHOD: We examined the effect of six collagen-based dressings on their ability to adsorb and inactivate different granulocyte proteases, plasmin, human neutrophil elastase (HLE), and matrix metalloproteases (MMP)-1, -2, -8, and -9, by an integrated approach including immunoelectron microscopy. RESULTS: We observed a reduction of the proteolytic activities of plasmin, HLE, and MMP-1, -2, -8, and -9, both on the biomaterial surface and in human chronic wound fluid. The most pronounced effect was observed in collagen-based dressings, with the highest content of native collagen networks resembling dermis structures. CONCLUSION: Our data suggest that this treatment strategy might be beneficial for the chronic wound environment, with the potential to promote improved wound healing.


Biocompatible Materials/pharmacology , Collagen/pharmacology , Dermis/drug effects , Diabetic Foot/therapy , Wound Healing , Biocompatible Materials/administration & dosage , Case-Control Studies , Collagen/administration & dosage , Dermis/ultrastructure , Diabetic Foot/enzymology , Diabetic Foot/pathology , Humans , Matrix Metalloproteinases/drug effects , Microscopy, Electron, Transmission , Occlusive Dressings , Peptide Hydrolases/drug effects
9.
J Am Soc Nephrol ; 28(8): 2472-2481, 2017 Aug.
Article En | MEDLINE | ID: mdl-28289183

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


Cell-Derived Microparticles/physiology , Complement C1 Inactivator Proteins/physiology , Vasculitis/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Chemotaxis , Child , Complement C1 Inhibitor Protein , Endothelium, Vascular/cytology , Female , Humans , Male , Middle Aged , Young Adult
11.
EBioMedicine ; 16: 302-311, 2017 Feb.
Article En | MEDLINE | ID: mdl-28139439

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.


Kidney Glomerulus/metabolism , Kidney/metabolism , Neutrophils/enzymology , Peptide Hydrolases/metabolism , Thrombosis/metabolism , von Willebrand Factor/metabolism , ADAMTS13 Protein/metabolism , Adult , Blood Platelets/metabolism , Cathepsin G/metabolism , Endothelial Cells/metabolism , Glomerular Basement Membrane/drug effects , Glomerular Basement Membrane/metabolism , Humans , Immunoblotting , Kidney/blood supply , Kidney/ultrastructure , Kidney Glomerulus/drug effects , Kidney Glomerulus/ultrastructure , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Microscopy, Electron, Transmission , Myeloblastin/metabolism , Proteinase Inhibitory Proteins, Secretory/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thrombosis/prevention & control
12.
J Intern Med ; 281(2): 123-148, 2017 02.
Article En | MEDLINE | ID: mdl-27723152

Haemolytic uraemic syndrome (HUS) is defined by the simultaneous occurrence of nonimmune haemolytic anaemia, thrombocytopenia and acute renal failure. This leads to the pathological lesion termed thrombotic microangiopathy, which mainly affects the kidney, as well as other organs. HUS is associated with endothelial cell injury and platelet activation, although the underlying cause may differ. Most cases of HUS are associated with gastrointestinal infection with Shiga toxin-producing enterohaemorrhagic Escherichia coli (EHEC) strains. Atypical HUS (aHUS) is associated with complement dysregulation due to mutations or autoantibodies. In this review, we will describe the causes of HUS. In addition, we will review the clinical, pathological, haematological and biochemical features, epidemiology and pathogenetic mechanisms as well as the biochemical, microbiological, immunological and genetic investigations leading to diagnosis. Understanding the underlying mechanisms of the different subtypes of HUS enables tailoring of appropriate treatment and management. To date, there is no specific treatment for EHEC-associated HUS but patients benefit from supportive care, whereas patients with aHUS are effectively treated with anti-C5 antibody to prevent recurrences, both before and after renal transplantation.


Hemolytic-Uremic Syndrome , Diagnosis, Differential , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/epidemiology , Hemolytic-Uremic Syndrome/physiopathology , Hemolytic-Uremic Syndrome/therapy , Humans , Prognosis
13.
Kidney Int ; 90(4): 726-9, 2016 10.
Article En | MEDLINE | ID: mdl-27633864

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 Mbls. Treatment with anti-human MBL2 antibody was protective, suggesting that MBL pathway activation also contributes to Shiga toxin-mediated renal injury.


Escherichia coli Infections , Hemolytic-Uremic Syndrome , Mannose-Binding Lectin , Shiga-Toxigenic Escherichia coli , Animals , Humans , Kidney , Mice , Shiga Toxin
14.
Adv Exp Med Biol ; 865: 19-42, 2015.
Article En | MEDLINE | ID: mdl-26306441

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


Complement Activation , Complement System Proteins/metabolism , Hemolytic-Uremic Syndrome/metabolism , Purpura, Thrombotic Thrombocytopenic/metabolism , Thrombosis/metabolism , Vasculitis/metabolism , Blood Coagulation Factors/immunology , Blood Coagulation Factors/metabolism , Blood Platelets/immunology , Blood Platelets/metabolism , Blood Platelets/pathology , Cell-Derived Microparticles/immunology , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Complement System Proteins/immunology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/pathology , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/pathology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Purpura, Thrombotic Thrombocytopenic/immunology , Purpura, Thrombotic Thrombocytopenic/pathology , Thrombosis/immunology , Thrombosis/pathology , Vasculitis/immunology , Vasculitis/pathology
15.
J Immunol ; 191(5): 2184-93, 2013 Sep 01.
Article En | MEDLINE | ID: mdl-23878316

This study addressed the contribution of ADAMTS13 deficiency to complement activation in thrombotic thrombocytopenic purpura (TTP). Renal tissue and blood samples were available from 12 TTP patients. C3 and C5b-9 deposition were demonstrated in the renal cortex of two TTP patients, by immunofluorescence and immunohistochemistry, respectively. C3 was also demonstrated in the glomeruli of Shiga toxin-2-treated Adamts13(-/-) mice (n = 6 of 7), but less in mice that were not Shiga toxin-2 treated (n = 1 of 8, p < 0.05) or wild-type mice (n = 0 of 7). TTP patient plasma (n = 9) contained significantly higher levels of complement-coated endothelial microparticles than control plasma (n = 13), as detected by flow cytometry. Exposure of histamine-stimulated primary glomerular endothelial cells to platelet-rich plasma from patients, or patient platelet-poor plasma combined with normal platelets, in a perfusion system, under shear, induced C3 deposition on von Willebrand factor-platelet strings (on both von Willebrand factor and platelets) and on endothelial cells. Complement activation occurred via the alternative pathway. No C3 was detected when cells were exposed to TTP plasma that was preincubated with EDTA or heat-inactivated, or to control plasma. In the perfusion system, patient plasma induced more release of C3- and C9-coated endothelial microparticles compared with control plasma. The results indicate that the microvascular process induced by ADAMTS13 deficiency triggers complement activation on platelets and the endothelium, which may contribute to formation of thrombotic microangiopathy.


ADAM Proteins/deficiency , Complement Activation/immunology , Thrombotic Microangiopathies/immunology , Thrombotic Microangiopathies/metabolism , ADAM Proteins/immunology , ADAMTS13 Protein , Animals , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Metalloendopeptidases/deficiency , Metalloendopeptidases/immunology , Mice , Mice, Knockout
16.
Nephrol Dial Transplant ; 26(10): 3399-403, 2011 Oct.
Article En | MEDLINE | ID: mdl-21810760

A 4-year-old boy presented with proteinuria and developed progressive renal failure over 6 years. In the patient's family, five individuals were affected with atypical haemolytic uraemic syndrome (aHUS) but not the patient. Renal biopsies (n = 3) showed glomerular basement membrane thickening with double contours, endothelial swelling and deposits of C3 and C1q. Electron microscopy revealed mesangial and subendothelial electron-dense deposits. Complement mutations in membrane cofactor protein (Y155D) and C3 (R713W and G1094R) were detected in all affected family members. The patient also had transient autoantibodies to factor H. The findings suggest that aHUS and glomerulopathy resembling membranoproliferative glomerulonephritis may have a common molecular background.


Glomerulonephritis, Membranoproliferative/diagnosis , Kidney Failure, Chronic/etiology , Proteinuria/complications , Thrombotic Microangiopathies/diagnosis , Child, Preschool , Complement C3/genetics , Diagnosis, Differential , Female , Glomerulonephritis, Membranoproliferative/etiology , Humans , Male , Membrane Cofactor Protein/genetics , Mutation/genetics , Pedigree , Polymerase Chain Reaction , Prognosis , Thrombotic Microangiopathies/etiology
17.
PLoS One ; 6(6): e21587, 2011.
Article En | MEDLINE | ID: mdl-21720563

BACKGROUND: 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. METHODOLOGY/PRINCIPAL FINDINGS: 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. CONCLUSIONS/SIGNIFICANCE: 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.


ADAM Proteins/metabolism , Endothelial Cells/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Metalloendopeptidases/metabolism , ADAMTS13 Protein , Animals , Blood Platelets/metabolism , Capillaries/metabolism , Cells, Cultured , Culture Media/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/ultrastructure , Matrix Metalloproteinase 9/metabolism , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Mice , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism
18.
Am J Pathol ; 176(2): 608-18, 2010 Feb.
Article En | MEDLINE | ID: mdl-20056836

IgA nephropathy (IgAN) and Henoch-Schönlein purpura (HSP) are diseases characterized by IgA deposits in the kidney and/or skin. Both may arise after upper respiratory tract infections, but the pathogenic mechanisms governing these diseases remain unclear. Patients with IgAN (n = 16) and HSP (n = 17) were included in this study aimed at examining whether IgA-binding M proteins of group A streptococci could be involved. As M proteins vary in sequence, the study focused on the IgA-binding-region (IgA-BR) of three different M proteins: M4, M22, and M60. Renal tissue from IgAN and HSP patients and skin from HSP patients were examined for deposits of streptococcal IgA-BR by immunohistochemistry and electron microscopy using specific antibodies, and a skin sample from a HSP patient was examined by mass spectrometry. IgA-BR deposits were detected in 10/16 IgAN kidneys and 7/13 HSP kidneys. Electron microscopy demonstrated deposits of IgA-BRs in the mesangial matrix and glomerular basement membrane, which colocalized with IgA. Skin samples exhibited IgA-BR deposits in 4/5 biopsies, a result confirmed by mass spectrometry in one patient. IgA-BR deposits were not detected in normal kidney and skin samples. Taken together, these results demonstrate IgA-BR from streptococcal M proteins in patient tissues. IgA-BR, would on gaining access to the circulation, encounter circulatory IgA and form a complex with IgA-Fc that could deposit in tissues and contribute to the pathogenesis of IgAN and HSP.


Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Glomerulonephritis, IGA/metabolism , IgA Vasculitis/metabolism , Immunoglobulin A/metabolism , Precipitins/metabolism , Adolescent , Adult , Amino Acid Sequence , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Biopsy , Carrier Proteins/chemistry , Carrier Proteins/immunology , Child , Child, Preschool , Female , Glomerulonephritis, IGA/pathology , Humans , IgA Vasculitis/pathology , Immunohistochemistry , Kidney/metabolism , Kidney/pathology , Male , Microscopy, Electron , Molecular Sequence Data , Precipitins/ultrastructure , Protein Binding , Skin/metabolism , Skin/pathology , Skin/ultrastructure , Young Adult
19.
Pediatr Nephrol ; 25(1): 87-96, 2010 Jan.
Article En | MEDLINE | ID: mdl-19644711

ADAMTS13 mRNA, which encodes the von Willebrand factor-cleaving protease, has been detected in a variety of tissues, including the kidney. The aim of our study was to characterize tubular expression and bioactivity of ADAMTS13. ADAMTS13 mRNA was detected in cultured primary human renal tubular epithelial cells (HRTEC) and in A498 cells, a human renal carcinoma cell line, by real-time PCR. Protein was detected using immunofluorescence and immunoblotting. Immunoblots demonstrated that the protein was secreted. The protease was proteolytically active in both cell lysates and cleaved the FRETS­VWF73 substrate. ADAMTS13 was demonstrated in situ in the renal cortex by immunohistochemistry. Protease was detected in both the proximal and distal renal tubules in normal renal tissue (n=3) as well as in patients with tubular disorders (n=3). Immunoblotting revealed that ADAMTS13 was present in the urine of patients with tubulopathy (n=5) but not in normal urine. ADAMTS13 in urine had a molecular size similar to that in plasma, which indicates that the protease originates in the tubuli because such large proteins do not normally pass the glomerular filter. In conclusion, human renal tubular epithelial cells synthesize biologically active ADAMTS13 which may, after release from tubuli, regulate hemostasis in the local microenvironment.


ADAM Proteins/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , ADAM Proteins/genetics , ADAMTS13 Protein , Adolescent , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Child , Child, Preschool , Female , Gene Expression , Humans , Immunohistochemistry , Kidney Diseases/pathology , Kidney Diseases/urine , Kidney Neoplasms/metabolism , Kidney Tubules, Proximal/pathology , Male , RNA, Messenger/metabolism
20.
Am J Pathol ; 173(5): 1428-39, 2008 Nov.
Article En | MEDLINE | ID: mdl-18832584

Toll-like receptors (TLRs) are key factors of innate immunity that detect pathogen invasion and trigger a host response. TLR4 can mediate a response through adaptor molecules, MyD88 or TRIF. In the present study, streptomycin-treated MyD88(-/-), Tlr4(-/-), Trif (Lps2/Lps2), and C57BL/6 wild-type (WT) mice were infected with either Shiga toxin (Stx)-producing or non-producing Escherichia coli O157:H7. Moderate to severe clinical signs of disease developed in MyD88(-/-) (n = 21/21), Tlr4(-/-) (n = 12/16), Trif (Lps2/Lps2) (n = 7/15) and WT mice (n = 6/20) infected with Stx-producing E. coli O157:H7 but not in mice inoculated with the Stx non-producing strain (n = 0/54, P < 0.001). MyD88(-/-) mice infected with Stx-producing E. coli O157:H7 developed the most severe disease and had the highest bacterial burden. Hematological analysis of sick MyD88(-/-) mice showed reduced red blood cell counts and reticulocytosis, suggesting hemolysis. Thrombocytopenia developed in MyD88(-/-), Trif (Lps2/Lps2), and WT mice, and creatinine levels were elevated in both MyD88(-/-) and WT mice infected with the Stx-producing strain. Renal histopathology showed evidence of glomerular capillary congestion, tubular desquamation, and fibrinogen deposition, and intestinal histopathology showed mucosal injury, edema, and inflammation in sick mice. Administration of purified Stx2 to MyD88(-/-) and WT mice led to severe disease in both groups, suggesting that MyD88(-/-) mice are not more sensitive to Stx than WT mice. As MyD88(-/-) mice developed the most severe disease hematological and pathological changes, the results suggest that dysfunctional innate immune responses via MyD88 enhanced Stx-induced disease.


Escherichia coli Infections/microbiology , Escherichia coli O157/physiology , Myeloid Differentiation Factor 88/deficiency , Shiga Toxin/immunology , Anemia/microbiology , Animals , Blood Cell Count , Body Weight , Colony Count, Microbial , Creatinine/blood , Escherichia coli Infections/blood , Escherichia coli Infections/physiopathology , Escherichia coli O157/growth & development , Fibrinogen/metabolism , Hemolysis , Immunity, Innate , Intestines/microbiology , Intestines/pathology , Kidney/microbiology , Kidney/pathology , Kidney/physiopathology , Kidney Function Tests , Mice , Mice, Inbred C57BL , Models, Immunological , Myeloid Differentiation Factor 88/metabolism , Platelet Count , Shiga Toxin/isolation & purification , Toll-Like Receptors/metabolism
...