Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Clin Exp Rheumatol ; 40(11): 2153-2160, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35579081

ABSTRACT

OBJECTIVES: We aimed to determine the presence, amount and origin of microchimerism in peripheral blood of pregnant and non-pregnant parous women with systemic lupus erythematosus (SLE) as compared to control subjects. METHODS: We performed a comparative study in which peripheral blood was drawn from eleven female non-pregnant SLE-patients and 22 control subjects, and from six pregnant SLE-patients and eleven control subjects during gestation and up to six months postpartum. Quantitative PCR for insertion-deletion polymorphisms and null alleles was used to detect microchimerism in peripheral blood mononuclear cells and granulocytes. RESULTS: Microchimerism was detected more often in non-pregnant SLE-patients than control subjects (54.4% vs. 13.6%, respectively; p=0.03). When present, the median total number of foetal chimeric cells was 5 gEq/106 in patients and 2.5gEq/106 in control subjects (p=0.048). Microchimerism was mostly foetal in origin; maternal microchimerism was detected in one patient and one control subject. In control subjects, microchimerism was always derived from only one source whereas in 50% of patients it originated from multiple sources. The pregnant patients had a significantly higher median number of foetal chimeric cells in the granulocyte fraction just after delivery than control subjects (7.5 gEq/106 vs. 0 gEq/106, respectively; p=0.02). CONCLUSIONS: Just after delivery, SLE-patients had more microchimerism than control subjects. Three months post-partum, microchimerism was no longer detectable, only to reappear many years after the last pregnancy, more often and at higher levels in SLE-patients than in control subjects. This suggests that these chimeric cells may originate from non-circulating foetal chimeric stem cells.


Subject(s)
Lupus Erythematosus, Systemic , Pregnancy Complications , Pregnancy , Humans , Female , Chimerism , Leukocytes, Mononuclear , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/genetics , Real-Time Polymerase Chain Reaction
2.
Arterioscler Thromb Vasc Biol ; 37(7): 1371-1379, 2017 07.
Article in English | MEDLINE | ID: mdl-28495931

ABSTRACT

OBJECTIVE: An excessive release and impaired degradation of neutrophil extracellular traps (NETs) leads to the continuous exposure of NETs to the endothelium in a variety of hematologic and autoimmune disorders, including lupus nephritis. This study aims to unravel the mechanisms through which NETs jeopardize vascular integrity. APPROACH AND RESULTS: Microvascular and macrovascular endothelial cells were exposed to NETs, and subsequent effects on endothelial integrity and function were determined in vitro and in vivo. We found that endothelial cells have a limited capacity to internalize NETs via the receptor for advanced glycation endproducts. An overflow of the phagocytic capacity of endothelial cells for NETs resulted in the persistent extracellular presence of NETs, which rapidly altered endothelial cell-cell contacts and induced vascular leakage and transendothelial albumin passage through elastase-mediated proteolysis of the intercellular junction protein VE-cadherin. Furthermore, NET-associated elastase promoted the nuclear translocation of junctional ß-catenin and induced endothelial-to-mesenchymal transition in cultured endothelial cells. In vivo, NETs could be identified in kidney samples of diseased MRL/lpr mice and patients with lupus nephritis, in whom the glomerular presence of NETs correlated with the severity of proteinuria and with glomerular endothelial-to-mesenchymal transition. CONCLUSIONS: These results indicate that an excess of NETs exceeds the phagocytic capacity of endothelial cells for NETs and promotes vascular leakage and endothelial-to-mesenchymal transition through the degradation of VE-cadherin and the subsequent activation of ß-catenin signaling. Our data designate NET-associated elastase as a potential therapeutic target in the prevention of endothelial alterations in diseases characterized by aberrant NET release.


Subject(s)
Epithelial-Mesenchymal Transition , Extracellular Traps/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Kidney Glomerulus/metabolism , Lupus Nephritis/metabolism , Neutrophils/metabolism , Adult , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability , Clathrin/metabolism , Disease Models, Animal , Endocytosis , Extracellular Traps/immunology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Leukocyte Elastase/metabolism , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Mice, Inbred CBA , Mice, Inbred MRL lpr , Neutrophils/immunology , Neutrophils/pathology , Phagocytosis , Receptor for Advanced Glycation End Products/metabolism , Severity of Illness Index , Signal Transduction , Time Factors , Young Adult , beta Catenin/metabolism
3.
PLoS One ; 11(11): e0167336, 2016.
Article in English | MEDLINE | ID: mdl-27898729

ABSTRACT

Glycosaminoglycans (GAGs) are linear negatively charged polysaccharides and important components of extracellular matrices and cell surface glycan layers such as the endothelial glycocalyx. The GAG family includes sulfated heparin, heparan sulfate (HS), dermatan sulfate (DS), chondroitin sulfate (CS), keratan sulfate, and non-sulfated hyaluronan. Because relative expression of GAGs is dependent on cell-type and niche, isolating GAGs from cell cultures and tissues may provide insight into cell- and tissue-specific GAG structure and functions. In our objective to obtain structural information about the GAGs expressed on a specialized mouse glomerular endothelial cell culture (mGEnC-1) we adapted a recently published GAG isolation protocol, based on cell lysis, proteinase K and DNase I digestion. Analysis of the GAGs contributing to the mGEnC-1 glycocalyx indicated a large HS and a minor CS content on barium acetate gel. However, isolated GAGs appeared resistant to enzymatic digestion by heparinases. We found that these GAG extracts were heavily contaminated with RNA, which co-migrated with HS in barium acetate gel electrophoresis and interfered with 1,9-dimethylmethylene blue (DMMB) assays, resulting in an overestimation of GAG yields. We hypothesized that RNA may be contaminating GAG extracts from other cell cultures and possibly tissue, and therefore investigated potential RNA contaminations in GAG extracts from two additional cell lines, human umbilical vein endothelial cells and retinal pigmental epithelial cells, and mouse kidney, liver, spleen and heart tissue. GAG extracts from all examined cell lines and tissues contained varying amounts of contaminating RNA, which interfered with GAG quantification using DMMB assays and characterization of GAGs by barium acetate gel electrophoresis. We therefore recommend routinely evaluating the RNA content of GAG extracts and propose a robust protocol for GAG isolation that includes an RNA digestion step.


Subject(s)
Glycosaminoglycans/chemistry , Kidney/metabolism , Liver/metabolism , RNA/isolation & purification , Spleen/metabolism , Alginates/chemistry , Animals , Cell Line , Chondroitin Sulfates/chemistry , Dermatan Sulfate/chemistry , Electrophoresis, Agar Gel , Glucuronic Acid/chemistry , Glycosaminoglycans/isolation & purification , Glycosaminoglycans/standards , Heparitin Sulfate/chemistry , Hexuronic Acids/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronic Acid/chemistry , Keratan Sulfate/chemistry , Mice , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism
4.
Kidney Int ; 90(5): 1012-1022, 2016 11.
Article in English | MEDLINE | ID: mdl-27575559

ABSTRACT

Proteinuria is one of the first clinical signs of diabetic nephropathy and an independent predictor for the progression to renal failure. Cathepsin L, a lysosomal cysteine protease, can be involved in the development of proteinuria by degradation of proteins that are important for normal podocyte architecture, such as the CD2-associated protein, synaptopodin, and dynamin. Cathepsin L also activates heparanase, a heparan sulfate endoglycosidase previously shown to be crucial for the development of diabetic nephropathy. Here, we evaluated the exact mode of action of cathepsin L in the development of proteinuria in streptozotocin-induced diabetes. Cathepsin L-deficient mice, in contrast to their wild-type littermates, failed to develop albuminuria, mesangial matrix expansion, tubulointerstitial fibrosis, and renal macrophage influx and showed a normal renal function. In wild-type mice the early development of albuminuria correlated with the activation of heparanase and loss of heparan sulfate expression, whereas loss of synaptopodin expression and podocyte damage occurred at a later stage. Thus, cathepsin L is causally involved in the pathogenesis of experimental diabetic nephropathy. Most likely, cathepsin L-dependent heparanase activation is crucial for the development of albuminuria and renal damage.


Subject(s)
Cathepsin L/metabolism , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cytoskeletal Proteins/metabolism , Dynamins/metabolism , Glucuronidase/metabolism , Mice, Inbred C57BL , Microfilament Proteins/metabolism
5.
J Am Soc Nephrol ; 27(12): 3545-3551, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27026367

ABSTRACT

Diabetic nephropathy (DN) is the leading cause of CKD in the Western world. Endothelin receptor antagonists have emerged as a novel treatment for DN, but the mechanisms underlying the protective effect remain unknown. We previously showed that both heparanase and endothelin-1 are essential for the development of DN. Here, we further investigated the role of these proteins in DN, and demonstrated that endothelin-1 activates podocytes to release heparanase. Furthermore, conditioned podocyte culture medium increased glomerular transendothelial albumin passage in a heparanase-dependent manner. In mice, podocyte-specific knockout of the endothelin receptor prevented the diabetes-induced increase in glomerular heparanase expression, consequent reduction in heparan sulfate expression and endothelial glycocalyx thickness, and development of proteinuria observed in wild-type counterparts. Our data suggest that in diabetes, endothelin-1 signaling, as occurs in endothelial activation, induces heparanase expression in the podocyte, damage to the glycocalyx, proteinuria, and renal failure. Thus, prevention of these effects may constitute the mechanism of action of endothelin receptor blockers in DN.


Subject(s)
Endothelin-1/physiology , Glucuronidase/physiology , Glycocalyx/enzymology , Kidney Glomerulus/enzymology , Kidney Glomerulus/ultrastructure , Proteinuria/etiology , Animals , Diabetic Nephropathies/etiology , Male , Mice , Podocytes/enzymology
6.
Am J Pathol ; 186(4): 794-804, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26851346

ABSTRACT

Vitamin D plays an important role in renal (patho)physiology. Patients with glomerular diseases have an injured renal filtration barrier, leading to proteinuria and reduced renal function. An impaired renal function also leads to 1,25-vitamin D3 deficiency as a result of reduced renal 1α-hydroxylase activity. Vitamin D treatment to reduce proteinuria remains controversial, although there is an inverse correlation between vitamin D levels and proteinuria. Herein, we showed that 1,25-vitamin D3-deficient 25-hydroxy-vitamin-D3-1α-hydroxylase knockout mice and 1,25-vitamin D3-deficient rats develop podocyte injury and renal dysfunction. Glomerular injury was characterized by proteinuria and partial podocyte foot process effacement. Expression of nephrin, podocin, desmin, and transient receptor potential channel C6 in the podocyte was significantly altered in 1,25-vitamin D3-deficient animals. Supplementation with 1,25-vitamin D3 or 1,25-vitamin D2 prevented podocyte effacement or reversed glomerular and tubulointerstitial damage in 1,25-vitamin D3-deficient animals, thereby preserving and restoring renal function, respectively. The effect of 1,25-vitamin D3 deficiency and 1,25-vitamin D3 and 1,25-vitamin D2 repletion on proteinuria could not be explained by hypocalcemia, changes in parathyroid hormone, or fibroblast growth factor 23. This study demonstrates that 1,25-vitamin D3 deficiency directly leads to renal injury in rodents. Translated to human subjects, this would underline the need for early vitamin D supplementation in patients with glomerular disease and chronic renal insufficiency, which might inhibit or potentially reverse renal injury.


Subject(s)
Albuminuria/etiology , Albuminuria/metabolism , Cholecalciferol/deficiency , Kidney Diseases/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Animals , Kidney Glomerulus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Parathyroid Hormone/metabolism , Rats , Rats, Wistar
7.
Am J Pathol ; 186(4): 805-15, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26873445

ABSTRACT

Heparanase, a heparan sulfate (HS)--specific endoglucuronidase, mediates the onset of proteinuria and renal damage during experimental diabetic nephropathy. Glomerular heparanase expression is increased in most proteinuric diseases. Herein, we evaluated the role of heparanase in two models of experimental glomerulonephritis, being anti-glomerular basement membrane and lipopolysaccharide-induced glomerulonephritis, in wild-type and heparanase-deficient mice. Induction of experimental glomerulonephritis led to an increased heparanase expression in wild-type mice, which was associated with a decreased glomerular expression of a highly sulfated HS domain, and albuminuria. Albuminuria was reduced in the heparanase-deficient mice in both models of experimental glomerulonephritis, which was accompanied by a better renal function and less renal damage. Notably, glomerular HS expression was preserved in the heparanase-deficient mice. Glomerular leukocyte and macrophage influx was reduced in the heparanase-deficient mice, which was accompanied by a reduced expression of both types 1 and 2 helper T-cell cytokines. In vitro, tumor necrosis factor-α and lipopolysaccharide directly induced heparanase expression and increased transendothelial albumin passage. Our study shows that heparanase contributes to proteinuria and renal damage in experimental glomerulonephritis by decreasing glomerular HS expression, enhancing renal leukocyte and macrophage influx, and affecting the local cytokine milieu.


Subject(s)
Diabetic Nephropathies/metabolism , Glomerular Basement Membrane/metabolism , Glomerulonephritis/etiology , Glomerulonephritis/metabolism , Glucuronidase/metabolism , Acute Disease , Animals , Heparitin Sulfate/metabolism , Mice, Inbred C57BL , Proteinuria/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
PLoS One ; 10(9): e0134946, 2015.
Article in English | MEDLINE | ID: mdl-26322947

ABSTRACT

Dermatan sulfate (DS), also known as chondroitin sulfate (CS)-B, is a member of the linear polysaccharides called glycosaminoglycans (GAGs). The expression of CS/DS and DS proteoglycans is increased in several fibrotic renal diseases, including interstitial fibrosis, diabetic nephropathy, mesangial sclerosis and nephrosclerosis. Little, however, is known about structural alterations in DS in renal diseases. The aim of this study was to evaluate the renal expression of two different DS domains in renal transplant rejection and glomerular pathologies. DS expression was evaluated in normal renal tissue and in kidney biopsies obtained from patients with acute interstitial or vascular renal allograft rejection, patients with interstitial fibrosis and tubular atrophy (IF/TA), and from patients with focal segmental glomerulosclerosis (FSGS), membranous glomerulopathy (MGP) or systemic lupus erythematosus (SLE), using our unique specific anti-DS antibodies LKN1 and GD3A12. Expression of the 4/2,4-di-O-sulfated DS domain recognized by antibody LKN1 was decreased in the interstitium of transplant kidneys with IF/TA, which was accompanied by an increased expression of type I collagen, decorin and transforming growth factor beta (TGF-ß), while its expression was increased in the interstitium in FSGS, MGP and SLE. Importantly, all patients showed glomerular LKN1 staining in contrast to the controls. Expression of the IdoA-Gal-NAc4SDS domain recognized by GD3A12 was similar in controls and patients. Our data suggest a role for the DS domain recognized by antibody LKN1 in renal diseases with early fibrosis. Further research is required to delineate the exact role of different DS domains in renal fibrosis.


Subject(s)
Dermatan Sulfate/metabolism , Graft Rejection/metabolism , Kidney Diseases/metabolism , Kidney/metabolism , Adolescent , Adult , Aged , Child, Preschool , Collagen Type I/metabolism , Female , Graft Rejection/pathology , Humans , Kidney/pathology , Kidney Diseases/pathology , Kidney Transplantation , Male , Middle Aged , Transforming Growth Factor beta/metabolism , Young Adult
9.
J Pathol ; 237(4): 472-81, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26202309

ABSTRACT

The glomerular filtration barrier consists of podocytes, the glomerular basement membrane, and endothelial cells covered with a glycocalyx. Heparan sulphate (HS) in the glomerular filtration barrier is reduced in patients with proteinuria, which is associated with increased expression of the HS-degrading enzyme heparanase. Previously, we showed that heparanase is essential for the development of proteinuria in experimental diabetic nephropathy. Vitamin D supplementation reduces podocyte loss and proteinuria in vitro and in vivo. Therefore, we hypothesize that vitamin D reduces proteinuria by reducing glomerular heparanase. Adriamycin-exposed rats developed proteinuria and showed increased heparanase expression, which was reduced by 1,25-dihydroxyvitamin D3 (1,25-D3) treatment. In vitro, adriamycin increased heparanase mRNA in the podocyte, which could be corrected by 1,25-D3 treatment. In addition, 1,25-D3 treatment reduced transendothelial albumin passage after adriamycin stimulation. In line with these results, we showed direct binding of the vitamin D receptor to the heparanase promoter, and 1,25-D3 dose-dependently reduced heparanase promoter activity. Finally, 1,25-D3-deficient 25-hydroxy-1α-hydroxylase knockout mice developed proteinuria and showed increased heparanase, which was normalized by 1,25-D3 treatment. Our data suggest that the protective effect of vitamin D on the development of proteinuria is mediated by inhibiting heparanase expression in the podocyte.


Subject(s)
Calcitriol/pharmacology , Glucuronidase/metabolism , Podocytes/enzymology , Proteinuria/metabolism , Animals , Chromatin Immunoprecipitation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Heparitin Sulfate/metabolism , Mice , Mice, Knockout , Podocytes/drug effects , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
10.
Mol Immunol ; 63(2): 203-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25246018

ABSTRACT

Complement factor H (FH) systemically inhibits excessive complement activation in the microenvironment of host cells, but for instance not on microbes. This self-recognition is mediated by two binding sites that recognize distinctly sulfated heparan sulfate (HS) domains. The interaction with HS not only concentrates FH on host cells, but directly affects its activity, evoking novel models of conformational activation. Genetic aberrations in the HS-binding domains systemically disturb the protective function of FH, yet the resulting loss of complement control affects mainly ocular and renal tissues. Recent results suggest that the specific expression of HS domains in these tissues restricts the interaction of HS to a single binding site within FH. This lack of redundancy could predispose eyes and kidneys to complement-mediated damage, making HS a central determinant for FH-associated diseases.


Subject(s)
Complement Factor H/immunology , Heparitin Sulfate/immunology , Kidney Diseases/immunology , Complement Factor H/chemistry , Complement Factor H/metabolism , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Humans , Organ Specificity
11.
Nephrol Dial Transplant ; 30(4): 560-4, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24811231

ABSTRACT

Mycophenolic acid (MPA) has become the cornerstone in the treatment of lupus nephritis. However, response rates are still far from ideal in clinical trials. Uncertainty exists regarding the correct dosing of MPA, and the recommended doses vary between recently published guidelines. Side effects are an additional problem resulting in frequent dose reduction and possible suboptimal exposure.In this review, we discuss the large variability between patients in drug exposure to MPA and the evidence for a relationship between drug exposure and efficacy in lupus nephritis. Methods for drug monitoring of MPA are discussed, and based on the current literature, we suggest as potential target levels a pre-dose level of 3.0 mg/L and an area under the concentration-versus-time curve between 35 and 45 mg h/L.Therapeutic drug monitoring may improve response rates in lupus nephritis by preventing low exposure and at the same time may reduce unnecessary side effects in patients who have high drug exposure with standard dose MPA. We specifically advise assessment of MPA drug exposure early after start of treatment and before concluding that treatment with MPA has failed.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Drug Monitoring/statistics & numerical data , Lupus Nephritis/drug therapy , Mycophenolic Acid/therapeutic use , Humans , Lupus Nephritis/diagnosis
12.
PLoS One ; 9(7): e102065, 2014.
Article in English | MEDLINE | ID: mdl-25019165

ABSTRACT

BACKGROUND: Activating mutations in the Transient Receptor Potential channel C6 (TRPC6) cause autosomal dominant focal segmental glomerular sclerosis (FSGS). TRPC6 expression is upregulated in renal biopsies of patients with idiopathic membranous glomerulopathy (iMN) and animal models thereof. In iMN, disease progression is characterized by glomerulosclerosis. In addition, a context-dependent TRPC6 overexpression was recently suggested in complement-mediated podocyte injury in e.g. iMN. Hence, we hypothesized that genetic variants in TRPC6 might affect susceptibility to development or progression of iMN. METHODS & RESULTS: Genomic DNA was isolated from blood samples of 101 iMN patients and 292 controls. By direct sequencing of the entire TRPC6 gene, 13 single nucleotide polymorphisms (SNPs) were identified in the iMN cohort, two of which were causing an amino acid substitution (rs3802829; Pro15Ser and rs36111323, Ala404Val). No statistically significant differences in genotypes or allele frequencies between patients and controls were observed. Clinical outcome in patients was determined (remission n = 26, renal failure n = 46, persistent proteinuria n = 29, follow-up median 80 months {range 51-166}). The 13 identified SNPs showed no association with remission or renal failure. There were no differences in genotypes or allele frequencies between patients in remission and progressors. CONCLUSIONS: Our data suggest that TRPC6 polymorphisms do not affect susceptibility to iMN, or clinical outcome in iMN.


Subject(s)
Genetic Predisposition to Disease , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/physiopathology , Polymorphism, Single Nucleotide/genetics , TRPC Cation Channels/genetics , Adult , Amino Acid Substitution/genetics , Base Sequence , DNA Primers/genetics , Disease Progression , Gene Frequency , Genetic Association Studies , Humans , Middle Aged , Molecular Sequence Data , Protein Conformation , Sequence Analysis, DNA , TRPC Cation Channels/chemistry , TRPC6 Cation Channel
13.
Kidney Int ; 86(5): 932-42, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24759151

ABSTRACT

The glomerular endothelial glycocalyx is postulated to be an important modulator of permeability and inflammation. The glycocalyx consists of complex polysaccharides, the main functional constituent of which, heparan sulfate (HS), is synthesized and modified by multiple enzymes. The N-deacetylase-N-sulfotransferase (Ndst) enzymes initiate and dictate the modification process. Here we evaluated the effects of modulation of HS in the endothelial glycocalyx on albuminuria and glomerular leukocyte influx using mice deficient in endothelial and leukocyte Ndst1 (TEKCre+/Ndst1flox/flox). In these mice, glomerular expression of a specific HS domain was significantly decreased, whereas the expression of other HS domains was normal. In the endothelial glycocalyx, this specific HS structure was not associated with albuminuria or with changes in renal function. However, glomerular leukocyte influx was significantly reduced during antiglomerular basement membrane nephritis, which was associated with less glomerular injury and better renal function. In vitro decreased adhesion of wild-type and Ndst1-deficient granulocytes to Ndst1-silenced glomerular endothelial cells was found, accompanied by a decreased binding of chemokines and L-selectin. Thus, modulation of HS in the glomerular endothelial glycocalyx significantly reduced the inflammatory response in antiglomerular basement membrane nephritis.


Subject(s)
Anti-Glomerular Basement Membrane Disease/metabolism , Chemotaxis, Leukocyte , Endothelial Cells/metabolism , Glycocalyx/metabolism , Heparitin Sulfate/metabolism , Kidney Glomerulus/metabolism , Leukocytes/metabolism , Animals , Anti-Glomerular Basement Membrane Disease/genetics , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/physiopathology , Anti-Glomerular Basement Membrane Disease/prevention & control , Autoantibodies , Cell Adhesion , Cell Line , Chemokines/metabolism , Coculture Techniques , Disease Models, Animal , Down-Regulation , Endothelial Cells/immunology , Female , Glycocalyx/immunology , Kidney Glomerulus/immunology , Kidney Glomerulus/physiopathology , L-Selectin/metabolism , Leukocytes/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , Signal Transduction , Sulfotransferases/deficiency , Sulfotransferases/genetics , Time Factors , Transfection
14.
Am J Pathol ; 184(6): 1715-26, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24731445

ABSTRACT

Slit diaphragm and podocyte damage is crucial in the pathogenesis of proteinuria in diabetic nephropathy (DNP). Gain-of-function mutations in TRPC6, a slit diaphragm-associated ion channel, cause glomerulosclerosis; TRPC6 expression is increased in acquired glomerular disease. Hyperglycemia and high intrarenal angiotensin II (AngII) levels could contribute to podocyte injury in DNP. We determined whether glucose regulates TRPC6 expression and TRPC6-mediated Ca(2+) influx into the podocyte and whether these effects are AngII dependent. High glucose levels increased TRPC6 mRNA and protein expression in cultured podocytes; however, TRPC1 and TRPC5 mRNA expression was unaltered. AngII and inducing podocyte injury also specifically increased TRPC6 expression. Angiotensin receptor blockade and inhibition of local AngII production through angiotensin-converting enzyme inhibition prevented glucose-mediated increased TRPC6 expression. In addition, high glucose concentration pretreatment enhanced Ca(2+) influx in podocytes, which was prevented by concomitant angiotensin receptor blockade application and TRPC6 knockdown. Studies with a TRPC6 luciferase promoter construct demonstrated a glucose concentration-dependent effect on TRPC6 promoter activity. In vivo, podocyte TRPC6 protein expression was increased in proteinuric streptozotocin-induced diabetic rats. These data suggest that glucose can activate a local renin-angiotensin system in the podocyte, leading to increased TRPC6 expression, which enhances TRPC6-mediated Ca(2+) influx. Regulation of TRPC6 expression could be an important factor in podocyte injury due to chronic hyperglycemia and the antiproteinuric effect of angiotensin receptor blockade or angiotensin-converting enzyme inhibition in DNP.


Subject(s)
Angiotensin II/metabolism , Diabetic Nephropathies/metabolism , Gene Expression Regulation , Glucose/metabolism , Podocytes/metabolism , TRPC Cation Channels/biosynthesis , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Mice , Mice, Knockout , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Podocytes/pathology , Rats , Rats, Wistar , Renin-Angiotensin System/genetics , TRPC Cation Channels/genetics , TRPC6 Cation Channel
15.
Nephrol Dial Transplant ; 29(1): 49-55, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166469

ABSTRACT

Proteinuria is a hallmark of many glomerular diseases and an independent risk factor for the progression of renal failure. Proteinuria results from damage to the glomerular filtration barrier (GFB), which plays a critical role in size- and charge-selective filtration. The GFB consists of three layers, which is the fenestrated endothelium that is covered by the glycocalyx, the podocytes and the intervening glomerular basement membrane. Defects in one of the three layers in the GFB can lead to the development of proteinuria. Heparan sulphate (HS) is a negatively charged polysaccharide that is abundantly expressed in all layers of the GFB. HS expression in the GFB is reduced in the majority of patients with proteinuria, which is associated with an increased glomerular expression of the HS-degrading enzyme heparanase. The primary role of HS in the development of proteinuria has been challenged after the establishment of several genetically engineered mouse models with an altered HS expression that did not display development of overt proteinuria. However, in a recent study, we showed that heparanase is essential for the development of proteinuria in diabetic nephropathy, which suggests that loss of HS contributes to the development of proteinuria. Recent studies also further highlight the importance of the glomerular endothelial glycocalyx in charge-selective filtration and the development of proteinuria. This review aims to summarize our current knowledge on the role of in particular HS and heparanase in the development of proteinuria.


Subject(s)
Glucuronidase/physiology , Glycocalyx/physiology , Animals , Diabetic Nephropathies/metabolism , Female , Glomerular Basement Membrane/metabolism , Heparitin Sulfate/metabolism , Humans , Kidney Glomerulus/metabolism , Male , Podocytes/metabolism , Proteinuria/etiology , Proteinuria/physiopathology
16.
Am J Pathol ; 183(5): 1571-1584, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24035513

ABSTRACT

Depending on the glycan structure, proteoglycans can act as coreceptors for growth factors. We hypothesized that proteoglycans and their growth factor ligands orchestrate tissue remodeling in chronic transplant dysfunction. We have previously shown perlecan to be selectively up-regulated in the glomeruli and arteries in a rat renal transplantation model. Using the same model, here we present quantitative RT-PCR profiling data on proteoglycans and growth factors from laser-microdissected glomeruli, arterial tunicae mediae, and neointimae at 12 weeks after transplantation. In glomeruli and neointimae of allografts, selective induction of the matrix heparan sulfate proteoglycan perlecan was observed, along with massive accumulation of fibroblast growth factor 2 (FGF2). Profiling the heparan sulfate polysaccharide side chains revealed conversion from a non-FGF2-binding heparan sulfate phenotype in control and isografted kidneys toward a FGF2-binding phenotype in allografts. In vitro experiments with perlecan-positive rat mesangial cells showed that FGF2-induced proliferation is dependent on sulfation and can be inhibited by exogenously added heparan sulfate. These findings indicate that matrix proteoglycans such as perlecan serve as functional docking platforms for FGF2 in chronic transplant dysfunction. We speculate that heparin-like glycomimetics could be a promising intervention to retard development of glomerulosclerosis and neointima formation in chronic transplant dysfunction.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Heparitin Sulfate/metabolism , Kidney Transplantation/adverse effects , Kidney/metabolism , Kidney/pathology , Proteoglycans/metabolism , Signal Transduction , Allografts/metabolism , Allografts/pathology , Amino Acid Motifs , Animals , Cell Membrane/metabolism , Cell Proliferation , Chronic Disease , Female , Heparan Sulfate Proteoglycans/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mesangial Cells/metabolism , Mesangial Cells/pathology , Protein Binding , Rats , Up-Regulation
17.
Am J Pathol ; 182(4): 1196-204, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23385000

ABSTRACT

The transient receptor potential cation channel C6 (TRPC6) is a slit diaphragm protein expressed by podocytes. TRPC6 gain-of-function mutations cause autosomal dominant focal segmental glomerulosclerosis. In acquired proteinuric renal disease, glomerular TRPC6 expression is increased. We previously demonstrated that acquired increased TRPC6 expression is ameliorated by antiproteinuric angiotensin receptor blockers and angiotensin-converting enzyme inhibitors. Vitamin D also has an antiproteinuric effect. We hypothesized that vitamin D reduces proteinuria by affecting TRPC6 expression in podocytes. Adriamycin-induced nephropathy increased TRPC6 mRNA and protein expression and induced proteinuria in rats. Treatment with 1,25-dihydroxyvitamin D3 (1,25-D3) normalized TRPC6 expression and reduced proteinuria. In vitro, podocyte injury induced by adriamycin exposure in cultured podocytes increased TRPC6 expression. Treatment of injured podocytes with 1,25-D3 dose dependently reduced adriamycin-induced TRPC6 expression. Chromatin immunoprecipitation analysis demonstrated that the vitamin D receptor directly binds to the TRPC6 promoter. Moreover, 1,25-D3 reduced TRPC6 promoter activity in a luciferase reporter assay. In 1,25-D3-deficient 25-hydroxy-1α-hydroxylase knockout mice, TRPC6 expression was increased, accompanied by podocyte foot process effacement and proteinuria. 1,25-D3 supplementation normalized TRPC6 expression, podocyte morphology, and proteinuria in these mice. These results demonstrate that vitamin D down-regulates the enhanced TRPC6 expression in in vivo and in vitro podocyte injury, possibly through a direct effect on TRPC6 promoter activity. This TRPC6 down-regulation could contribute to the antiproteinuric effect of vitamin D.


Subject(s)
Down-Regulation/drug effects , Kidney Diseases/pathology , Podocytes/metabolism , Podocytes/pathology , Proteinuria/pathology , TRPC Cation Channels/genetics , Vitamin D/pharmacology , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism , Animals , Chromatin Immunoprecipitation , Down-Regulation/genetics , Glomerulosclerosis, Focal Segmental/complications , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Diseases/complications , Kidney Diseases/genetics , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Mice , Podocytes/drug effects , Podocytes/ultrastructure , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Proteinuria/complications , Proteinuria/genetics , Rats , Rats, Wistar , Receptors, Calcitriol/metabolism , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , Vitamin D/analogs & derivatives
18.
Nephrol Dial Transplant ; 28(7): 1830-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23291369

ABSTRACT

BACKGROUND: Focal segmental glomerulosclerosis (FSGS) is a leading cause of steroid-resistant nephrotic syndrome. Hereditary FSGS is frequently caused by mutations in important structural podocyte proteins, including the slit diaphragm-associated transient receptor potential channel C6 (TRPC6). METHODS: In five patients with biopsy-proven autosomal-dominant FSGS from five different Dutch families, all 13 exons of TRPC6 were sequenced. Upon identification of a novel TRPC6 sequence variant, the resultant amino acid change was introduced in the wild-type TRPC6 protein and functionally tested using patch-clamp analyses and cell-surface biotinylation experiments. RESULTS: None of the previously described TRPC6 mutations were found in our cohort. In one family, we identified a novel c.524G>A sequence variant resulting in a p.Arg175Gln (R175Q) substitution in the TRPC6 protein. This sequence variant was absent in 449 control subjects and from public SNP databases. The mutation was located in the third ankyrin repeat domain (ANK3) in the cytoplasmic N-tail of TRPC6, important for protein-protein interaction and regulation of ion channel activity. Patch-clamp analyses of the mutant channel indeed showed an increased TRPC6 channel-mediated current. However, cell-surface expression of the mutant channel was not increased. CONCLUSIONS: We identified a novel TRPC6 p.Arg175Gln gain-of-function mutation that shows increased TRPC6-mediated current, which is not due to altered cell-surface expression. This is the first mutation identified in ANK3 of the TRPC6 N-tail and is most likely responsible for the late-onset autosomal dominant FSGS in this family.


Subject(s)
Consanguinity , Glomerulosclerosis, Focal Segmental/etiology , Mutation/genetics , TRPC Cation Channels/genetics , Adult , Age of Onset , Aged , Amino Acid Sequence , Child, Preschool , Electrophysiology , Family , Female , Follow-Up Studies , Glomerular Filtration Rate , HEK293 Cells , Humans , Male , Middle Aged , Molecular Sequence Data , Netherlands , Pedigree , Prognosis , Sequence Homology, Amino Acid , TRPC6 Cation Channel , Time Factors
19.
Ann Rheum Dis ; 71(11): 1771-82, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22851469

ABSTRACT

OBJECTIVES: To develop recommendations for the management of adult and paediatric lupus nephritis (LN). METHODS: The available evidence was systematically reviewed using the PubMed database. A modified Delphi method was used to compile questions, elicit expert opinions and reach consensus. RESULTS: Immunosuppressive treatment should be guided by renal biopsy, and aiming for complete renal response (proteinuria <0.5 g/24 h with normal or near-normal renal function). Hydroxychloroquine is recommended for all patients with LN. Because of a more favourable efficacy/toxicity ratio, as initial treatment for patients with class III-IV(A) or (A/C) (±V) LN according to the International Society of Nephrology/Renal Pathology Society 2003 classification, mycophenolic acid (MPA) or low-dose intravenous cyclophosphamide (CY) in combination with glucocorticoids is recommended. In patients with adverse clinical or histological features, CY can be prescribed at higher doses, while azathioprine is an alternative for milder cases. For pure class V LN with nephrotic-range proteinuria, MPA in combination with oral glucocorticoids is recommended as initial treatment. In patients improving after initial treatment, subsequent immunosuppression with MPA or azathioprine is recommended for at least 3 years; in such cases, initial treatment with MPA should be followed by MPA. For MPA or CY failures, switching to the other agent, or to rituximab, is the suggested course of action. In anticipation of pregnancy, patients should be switched to appropriate medications without reducing the intensity of treatment. There is no evidence to suggest that management of LN should differ in children versus adults. CONCLUSIONS: Recommendations for the management of LN were developed using an evidence-based approach followed by expert consensus.


Subject(s)
Azathioprine/therapeutic use , Cyclophosphamide/therapeutic use , Disease Management , Glucocorticoids/therapeutic use , Hydroxychloroquine/therapeutic use , Immunosuppressive Agents/therapeutic use , Lupus Nephritis/drug therapy , Mycophenolic Acid/therapeutic use , Adult , Biopsy , Child , Dose-Response Relationship, Drug , Drug Substitution , Drug Therapy, Combination , Evidence-Based Medicine , Female , Humans , Kidney/drug effects , Kidney/pathology , Lupus Nephritis/diagnosis , Lupus Nephritis/urine , Male , Pregnancy
20.
Nephrol Dial Transplant ; 27(7): 2853-61, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22187315

ABSTRACT

BACKGROUND: A reduced heparan sulphate (HS) expression in the glomerular basement membrane of patients with overt diabetic nephropathy is associated with an increased glomerular heparanase expression. We investigated the possible association of urinary heparanase activity with the development of proteinuria in patients with Type 1 diabetes (T1D), Type 2 diabetes (T2D), or membranous glomerulopathy (MGP) as non-diabetic disease controls. METHODS: Heparanase activity, albumin, HS and creatinine were measured in the urine of patients with T1D (n=58) or T2D (n=31), in patients with MGP (n=52) and in healthy controls (n=10). Heparanase messenger RNA (mRNA) expression in leukocytes was determined in a subgroup of patients with T1D (n=19). RESULTS: Urinary heparanase activity was increased in patients with T1D and T2D, which was more prominent in patients with macroalbuminuria, whereas no activity could be detected in healthy controls. Albuminuria levels were associated with increased urinary heparanase activity in diabetic patients (r=0.20; P<0.05) but not in patients with MGP (r=0.11; P=0.43). A lower urinary heparanase activity was observed in diabetic patients treated with inhibitors of the renin-angiotensin-aldosterone system (RAAS), when compared to diabetic patients treated with other anti-hypertensives. Additionally, urinary heparanase activity was associated with age in T1D and MGP. In MGP, heparanase activity and ß2-microglobulin excretion correlated. In patients with T1D, no differences in heparanase mRNA expression in leukocytes could be observed. CONCLUSIONS: Urinary heparanase activity is increased in diabetic patients with proteinuria. However, whether increased heparanase activity is a cause or consequence of proteinuria requires additional research.


Subject(s)
Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/urine , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/urine , Glomerular Basement Membrane/pathology , Glucuronidase/urine , Heparitin Sulfate/metabolism , Adult , Aged , Albuminuria/diagnosis , Blotting, Western , Case-Control Studies , Diabetes Complications/enzymology , Diabetes Complications/etiology , Diabetes Complications/urine , Female , Follow-Up Studies , Glucuronidase/genetics , Humans , Immunoenzyme Techniques , Male , Middle Aged , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Renin-Angiotensin System , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...