Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 62
1.
J Cell Biochem ; 123(5): 893-905, 2022 05.
Article En | MEDLINE | ID: mdl-35312102

Glucocerebrosidase (GCase), encoded by the GBA gene, degrades the ubiquitous glycosphingolipid glucosylceramide. Inherited GCase deficiency causes Gaucher disease (GD). In addition, carriers of an abnormal GBA allele are at increased risk for Parkinson's disease. GCase undergoes extensive modification of its four N-glycans en route to and inside the lysosome that is reflected in changes in molecular weight as detected with sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Fluorescent activity-based probes (ABPs) that covalently label GCase in reaction-based manner in vivo and in vitro allow sensitive visualization of GCase molecules. Using these ABPs, we studied the life cycle of GCase in cultured fibroblasts and macrophage-like RAW264.7 cells. Specific attention was paid to the impact of 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) supplementation to bicarbonate-buffered medium. Here, we report how HEPES-buffered medium markedly influences processing of GCase, its lysosomal degradation, and the total cellular enzyme level. HEPES-containing medium was also found to reduce maturation of other lysosomal enzymes (α-glucosidase and ß-glucuronidase) in cells. The presence of HEPES in bicarbonate containing medium increases GCase activity in GD-patient derived fibroblasts, illustrating how the supplementation of HEPES complicates the use of cultured cells for diagnosing GD.


Gaucher Disease , Glucosylceramidase , Bicarbonates/metabolism , Gaucher Disease/genetics , Gaucher Disease/metabolism , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , HEPES/metabolism , Humans , Lysosomes/metabolism
2.
Clin Transl Med ; 11(11): e558, 2021 11.
Article En | MEDLINE | ID: mdl-34841686

BACKGROUND: Improved understanding of the interconnectedness of structural remodeling processes in atrial fibrillation (AF) in patients could identify targets for future therapies. METHODS: We present transcriptome sequencing of atrial tissues of patients without AF, with paroxysmal AF, and persistent AF (total n = 64). RNA expression levels were validated in the same and an independent cohort with qPCR. Biological processes were assessed with histological and immunohistochemical analyses. RESULTS: In AF patients, epicardial cell gene expression decreased, contrasting with an upregulation of epithelial-to-mesenchymal transition (EMT) and mesenchymal cell gene expression. Immunohistochemistry demonstrated thickening of the epicardium and an increased proportion of (myo)fibroblast-like cells in the myocardium, supporting enhanced EMT in AF. We furthermore report an upregulation of endothelial cell proliferation, angiogenesis, and endothelial signaling. EMT and endothelial cell proliferation concurred with increased interstitial (myo)fibroblast-like cells and extracellular matrix gene expression including enhanced tenascin-C, thrombospondins, biglycan, and versican. Morphological analyses discovered increased and redistributed glycosaminoglycans and collagens in the atria of AF patients. Signaling pathways, including cell-matrix interactions, PI3K-AKT, and Notch signaling that could regulate mesenchymal cell activation, were upregulated. CONCLUSION: Our results suggest that EMT and endothelial cell proliferation work in concert and characterize the (myo)fibroblast recruitment and ECM remodeling of AF. These processes could guide future research toward the discovery of targets for AF therapy.


Atrial Fibrillation/complications , Endothelium/drug effects , Extracellular Matrix/physiology , Pericardium/drug effects , Aged , Atrial Fibrillation/physiopathology , Endothelium/metabolism , Extracellular Matrix/drug effects , Female , Fibroblasts/metabolism , Humans , Male , Middle Aged , Pericardium/metabolism
3.
JIMD Rep ; 62(1): 15-21, 2021 Nov.
Article En | MEDLINE | ID: mdl-34765393

Acid sphingomyelinase deficiency (ASMD) is a lysosomal storage disease (LSD) in which sphingomyelin accumulates due to deficient acid sphingomyelinase. In the chronic visceral subtype, organ manifestations are generally limited to the spleen, liver, and lungs. We report a male patient with the chronic visceral subtype who developed proteinuria and renal insufficiency at the age of 49. In renal tissue, foam cells were observed in the glomeruli as well as sphingomyelin accumulation within podocytes, mesangial cells, endothelial cells, and tubular epithelial cells. Although macrophages are the primary storage cells in both ASMD and Gaucher disease, comparison to the histopathological findings in Gaucher and Fabry disease revealed a diffuse storage pattern in multiple renal cell types, closer resembling the pattern found in Fabry disease.

4.
Physiol Rep ; 9(17): e15019, 2021 09.
Article En | MEDLINE | ID: mdl-34472715

Vascular endothelial cells are covered with glycocalyx comprising heparan sulfate, hyaluronan, chondroitin sulfate, and associated proteins. Glomerular endothelial glycocalyx is involved in protecting against induction of proteinuria and structural damage, but the specific components in glycocalyx that represent therapeutic targets remain unclear. Anti-vascular endothelial growth factor (VEGF) therapy is associated with an increased risk of glomerular endothelial injury. This study investigated whether hyaluronan could provide a therapeutic target to protect against proteinuria. We conducted ex vivo and in vivo experiments to explore the effects of degrading glomerular hyaluronan by administering hyaluronidase and of supplementation with hyaluronan. We investigated hyaluronan expression using biotin-labeled hyaluronan-binding protein (HABP) in human kidney specimens or serum hyaluronan in endothelial injuries under inhibition of VEGF signaling. We directly demonstrated hyaluronan in glomerular endothelial layers using HABP staining. Ex vivo and in vivo experiments showed the development of proteinuria after digestion of hyaluronan in glomerular capillaries. Supplementation with hyaluronan after hyaluronidase treatment suppressed proteinuria. Mice in the in vivo study developed albuminuria after intraperitoneal injection of hyaluronidase with decreased glomerular hyaluronan and increased serum hyaluronan. In human kidneys with endothelial cell dysfunction and proteinuria due to inhibition of VEGF, glomerular expression of hyaluronan was reduced even in normal-appearing glomeruli. Serum hyaluronan levels were elevated in patients with pre-eclampsia with VEGF signaling inhibition. Our data suggest that hyaluronan itself plays crucial roles in preventing proteinuria and preserving the integrity of endothelial cells. Hyaluronan could provide a therapeutic target for preventing glomerular endothelial glycocalyx damage, including VEGF signaling inhibition.


Endothelial Cells/metabolism , Glycocalyx/metabolism , Hyaluronic Acid/biosynthesis , Kidney Glomerulus/metabolism , Proteinuria/metabolism , Animals , Cattle , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Glycocalyx/drug effects , Glycocalyx/pathology , Humans , Hyaluronoglucosaminidase/administration & dosage , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Pregnancy , Proteinuria/pathology , Rats , Rats, Inbred Lew
5.
Clin Exp Nephrol ; 25(9): 1035-1046, 2021 Sep.
Article En | MEDLINE | ID: mdl-33999275

BACKGROUND: During peritoneal dialysis (PD), solute transport and ultrafiltration are mainly achieved by the peritoneal blood vasculature. Glycocalyx lies on the surface of endothelial cells and plays a role in vascular permeability. Low-glucose degradation product (GDP), pH-neutral PD solutions reportedly offer higher biocompatibility and lead to less peritoneal injury. However, the effects on the vasculature have not been clarified. METHODS: Peritoneal tissues from 11 patients treated with conventional acidic solutions (acidic group) and 11 patients treated with low-GDP, pH-neutral solutions (neutral group) were examined. Control tissues were acquired from 5 healthy donors of kidney transplants (control group). CD31 and ratio of luminal diameter to vessel diameter (L/V ratio) were evaluated to identify endothelial cells and vasculopathy, respectively. Immunostaining for heparan sulfate (HS) domains and Ulex europaeus agglutinin-1 (UEA-1) binding was performed to assess sulfated glycosaminoglycans and the fucose-containing sugar chain of glycocalyx. RESULTS: Compared with the acidic group, the neutral group showed higher CD31 positivity. L/V ratio was significantly higher in the neutral group, suggesting less progression of vasculopathy. Both HS expression and UEA-1 binding were higher in the neutral group, whereas HS expression was markedly more preserved than UEA-1 binding in the acidic group. In vessels with low L/V ratio, which were found only in the acidic group, HS expression and UEA-1 binding were diminished, suggesting a loss of glycocalyx. CONCLUSION: Peritoneal endothelial glycocalyx was more preserved in patients treated with low-GDP, pH-neutral solution. The use of low-GDP, pH-neutral solutions could help to protect peritoneal vascular structures and functions.


Capillaries/pathology , Dialysis Solutions/adverse effects , Endothelial Cells/metabolism , Glycocalyx/metabolism , Peritoneal Dialysis , Peritoneum/metabolism , Adult , Aged , Biopsy , Capillaries/metabolism , Dialysis Solutions/chemistry , Endothelial Cells/pathology , Female , Glucose/metabolism , Glycocalyx/pathology , Heparitin Sulfate/metabolism , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Peritoneum/blood supply , Peritoneum/pathology , Plant Lectins/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
6.
Int J Mol Sci ; 22(5)2021 Mar 03.
Article En | MEDLINE | ID: mdl-33802460

The lysosomal storage disease Niemann-Pick type C (NPC) is caused by impaired cholesterol efflux from lysosomes, which is accompanied by secondary lysosomal accumulation of sphingomyelin and glucosylceramide (GlcCer). Similar to Gaucher disease (GD), patients deficient in glucocerebrosidase (GCase) degrading GlcCer, NPC patients show an elevated glucosylsphingosine and glucosylated cholesterol. In livers of mice lacking the lysosomal cholesterol efflux transporter NPC1, we investigated the expression of established biomarkers of lipid-laden macrophages of GD patients, their GCase status, and content on the cytosol facing glucosylceramidase GBA2 and lysosomal integral membrane protein type B (LIMP2), a transporter of newly formed GCase to lysosomes. Livers of 80-week-old Npc1-/- mice showed a partially reduced GCase protein and enzymatic activity. In contrast, GBA2 levels tended to be reciprocally increased with the GCase deficiency. In Npc1-/- liver, increased expression of lysosomal enzymes (cathepsin D, acid ceramidase) was observed as well as increased markers of lipid-stressed macrophages (GPNMB and galectin-3). Immunohistochemistry showed that the latter markers are expressed by lipid laden Kupffer cells. Earlier reported increase of LIMP2 in Npc1-/- liver was confirmed. Unexpectedly, immunohistochemistry showed that LIMP2 is particularly overexpressed in the hepatocytes of the Npc1-/- liver. LIMP2 in these hepatocytes seems not to only localize to (endo)lysosomes. The recent recognition that LIMP2 harbors a cholesterol channel prompts the speculation that LIMP2 in Npc1-/- hepatocytes might mediate export of cholesterol into the bile and thus protects the hepatocytes.


Glucosylceramidase/metabolism , Liver/metabolism , Lysosomal Membrane Proteins/metabolism , Niemann-Pick Disease, Type C/metabolism , Receptors, Scavenger/metabolism , Animals , Biological Transport/physiology , Cathepsin D/metabolism , Cell Line , Cell Line, Tumor , Gaucher Disease/metabolism , Glucosylceramides/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Humans , Lysosomes/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , Sphingomyelins/metabolism
7.
J Transl Med ; 19(1): 38, 2021 01 20.
Article En | MEDLINE | ID: mdl-33472641

BACKGROUND: By binding to negatively charged polysaccharides called glycosaminoglycans, sodium can be stored in the body-particularly in the skin-without concurrent water retention. Concordantly, individuals with changed glycosaminoglycan structure (e.g. type 1 diabetes (DM1) and hereditary multiple exostosis (HME) patients) may have altered sodium and water homeostasis. METHODS: We investigated responses to acute (30-min infusion) and chronic (1-week diet) sodium loading in 8 DM1 patients and 7 HME patients in comparison to 12 healthy controls. Blood samples, urine samples, and skin biopsies were taken to investigate glycosaminoglycan sulfation patterns and both systemic and cellular osmoregulatory responses. RESULTS: Hypertonic sodium infusion increased plasma sodium in all groups, but more in DM1 patients than in controls. High sodium diet increased expression of nuclear factor of activated t-cells 5 (NFAT5)-a transcription factor responsive to changes in osmolarity-and moderately sulfated heparan sulfate in skin of healthy controls. In HME patients, skin dermatan sulfate, rather than heparan sulfate, increased in response to high sodium diet, while in DM1 patients, no changes were observed. CONCLUSION: DM1 and HME patients show distinct osmoregulatory responses to sodium loading when comparing to controls with indications for reduced sodium storage capacity in DM1 patients, suggesting that intact glycosaminoglycan biosynthesis is important in sodium and water homeostasis. Trial registration These trials were registered with the Netherlands trial register with registration numbers: NTR4095 ( https://www.trialregister.nl/trial/3933 at 2013-07-29) and NTR4788 ( https://www.trialregister.nl/trial/4645 at 2014-09-12).


Glycosaminoglycans , Sodium , Cross-Over Studies , Heparitin Sulfate , Humans , Netherlands
8.
Hum Mol Genet ; 29(7): 1168-1179, 2020 05 08.
Article En | MEDLINE | ID: mdl-32160276

Glutaric aciduria type 1 (GA1) is an inborn error of lysine degradation characterized by a specific encephalopathy that is caused by toxic accumulation of lysine degradation intermediates. Substrate reduction through inhibition of DHTKD1, an enzyme upstream of the defective glutaryl-CoA dehydrogenase, has been investigated as a potential therapy, but revealed the existence of an alternative enzymatic source of glutaryl-CoA. Here, we show that loss of DHTKD1 in glutaryl-CoA dehydrogenase-deficient HEK-293 cells leads to a 2-fold decrease in the established GA1 clinical biomarker glutarylcarnitine and demonstrate that oxoglutarate dehydrogenase (OGDH) is responsible for this remaining glutarylcarnitine production. We furthermore show that DHTKD1 interacts with OGDH, dihydrolipoyl succinyltransferase and dihydrolipoamide dehydrogenase to form a hybrid 2-oxoglutaric and 2-oxoadipic acid dehydrogenase complex. In summary, 2-oxoadipic acid is a substrate for DHTKD1, but also for OGDH in a cell model system. The classical 2-oxoglutaric dehydrogenase complex can exist as a previously undiscovered hybrid containing DHTKD1 displaying improved kinetics towards 2-oxoadipic acid.


Acyl Coenzyme A/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/genetics , Glutaryl-CoA Dehydrogenase/deficiency , Ketoglutarate Dehydrogenase Complex/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , Cells, Cultured , Glutaryl-CoA Dehydrogenase/genetics , Glutaryl-CoA Dehydrogenase/metabolism , HEK293 Cells , Humans , Ketone Oxidoreductases/genetics , Substrate Specificity/genetics
9.
Transl Res ; 217: 23-32, 2020 03.
Article En | MEDLINE | ID: mdl-31883728

Type 1 diabetes patients are more prone to have hypertension than healthy individuals, possibly mediated by increased blood pressure (BP) sensitivity to high salt intake. The classical concept proposes that the kidney is central in salt-mediated BP rises, by insufficient renal sodium excretion leading to extracellular fluid volume expansion. Recent animal-derived findings, however, propose a causal role for disturbance of macrophage-mediated lymphangiogenesis. Its relevance for humans, specifically type 1 diabetes patients, is unknown. The present study aimed to assess responses of type 1 diabetes patients to a dietary salt load with regard to BP, extracellular fluid volume (using precise iohexol measurements), and CD163+ macrophage and lymphatic capillary density in skin biopsies. Also, macrophage expression of HLA-DR (a proinflammatory marker) and CD206 (an anti-inflammatory marker) was assessed. Type 1 diabetes patients (n = 8) showed a salt-sensitive BP increase without extracellular fluid volume expansion. Whereas healthy controls (n = 12), who had no BP increase, showed increased skin CD163+ and HLA-DR+ macrophages and dilation of lymphatic skin vasculature after the dietary salt load, these changes were absent (and in case of HLA-DR more heterogenic) in type 1 diabetes patients. In conclusion, we show that salt sensitivity in type 1 diabetes patients cannot be explained by the classical concept of extracellular fluid volume expansion. Rather, we open up a potential role for macrophages and the lymphatic system. Future studies on hypertension and diabetes need to scrutinize these phenomena.


Blood Pressure/physiology , Diabetes Mellitus, Type 1/physiopathology , Extracellular Fluid/physiology , Lymphatic Vessels/physiology , Macrophages/physiology , Skin/immunology , Sodium Chloride, Dietary/administration & dosage , Adolescent , Adult , Cross-Over Studies , Humans , Male , Prospective Studies , Skin/blood supply , Young Adult
10.
JCI Insight ; 4(21)2019 11 01.
Article En | MEDLINE | ID: mdl-31672939

Inflammation may play a role in the link between high salt intake and its deleterious consequences. However, it is unknown whether salt can induce proinflammatory priming of monocytes and macrophages in humans. We investigated the effects of salt on monocytes and macrophages in vitro and in vivo by performing a randomized crossover trial in which 11 healthy human subjects adhered to a 2-week low-salt and high-salt diet. We demonstrate that salt increases monocyte expression of CCR2, a chemokine receptor that mediates monocyte infiltration in inflammatory diseases. In line with this, we show a salt-induced increase of plasma MCP-1, transendothelial migration of monocytes, and skin macrophage density after high-salt diet. Macrophages demonstrate signs of an increased proinflammatory phenotype after salt exposure, as represented by boosted LPS-induced cytokine secretion of IL-6, TNF, and IL-10 in vitro, and by increased HLA-DR expression and decreased CD206 expression on skin macrophages after high-salt diet. Taken together, our data open up the possibility for inflammatory monocyte and macrophage responses as potential contributors to the deleterious effects of high salt intake.


Inflammation/metabolism , Monocytes/drug effects , Receptors, CCR2/metabolism , Sodium Chloride, Dietary/pharmacology , Adult , Cross-Over Studies , Cytokines/metabolism , Female , Humans , Male , Monocytes/metabolism , Sodium Chloride, Dietary/metabolism , Young Adult
11.
Sci Rep ; 9(1): 12175, 2019 08 21.
Article En | MEDLINE | ID: mdl-31434958

Lymphatic absorption in the peritoneal cavity may contribute to ultrafiltration failure in peritoneal dialysis (PD). Lymphatic vessels develop during PD-related peritoneal fibrosis. Connective tissue growth factor (CTGF, also called CCN2) is an important determinant of fibrotic tissue remodeling, but little is known about its possible involvement in lymphangiogenesis. In this study, we investigated the relationship between CTGF and peritoneal lymphangiogenesis. A positive correlation was observed between vascular endothelial growth factor-C (VEGF-C), a major lymphangiogenic growth factor, and the CTGF concentration in human PD effluents. CTGF expression was positively correlated with expression of lymphatic markers and VEGF-C in human peritoneal biopsies. We found a positive correlation between the increase in CTGF and the increase in VEGF-C in cultured human peritoneal mesothelial cells (HPMCs) treated with transforming growth factor-ß1 (TGF-ß1). The diaphragm is a central player in peritoneal lymphatic absorption. CTGF expression was also correlated with expression of VEGF-C and lymphatics in a rat diaphragmatic fibrosis model induced by chlorhexidine gluconate (CG). Furthermore, CTGF gene deletion reduced VEGF-C expression and peritoneal lymphangiogenesis in the mouse CG model. Inhibition of CTGF also reduced VEGF-C upregulation in HPMCs treated with TGF-ß1. Our results suggest a close relationship between CTGF and PD-associated lymphangiogenesis.


Connective Tissue Growth Factor/metabolism , Lymphangiogenesis/physiology , Lymphatic Vessels/metabolism , Animals , Cell Line , Chlorhexidine/analogs & derivatives , Chlorhexidine/toxicity , Connective Tissue Growth Factor/antagonists & inhibitors , Connective Tissue Growth Factor/genetics , Disease Models, Animal , Humans , Lymphatic Vessels/pathology , Male , Mice , Peritoneal Dialysis , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Peritoneum/metabolism , Peritoneum/pathology , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism
12.
J Pathol ; 246(4): 470-484, 2018 12.
Article En | MEDLINE | ID: mdl-30073645

Obesity and dyslipidaemia are features of the metabolic syndrome and risk factors for chronic kidney disease. The cellular mechanisms connecting metabolic syndrome with chronic kidney disease onset and progression remain largely unclear. We show that proximal tubular epithelium is a target site for lipid deposition upon overnutrition with a cholesterol-rich Western-type diet. Affected proximal tubule epithelial cells displayed giant vacuoles of lysosomal or autophagosomal origin, harbouring oxidised lipoproteins and concentric membrane layer structures (multilamellar bodies), reminiscent of lysosomal storage diseases. Additionally, lipidomic analysis revealed renal deposition of cholesterol and phospholipids, including lysosomal phospholipids. Proteomic profiles of renal multilamellar bodies were distinct from those of epidermis or lung multilamellar bodies and of cytoplasmic lipid droplets. Tubular multilamellar bodies were observed in kidney biopsies of obese hypercholesterolaemic patients, and the concentration of the phospholipidosis marker di-docosahexaenoyl (22:6)-bis(monoacylglycerol) phosphate was doubled in urine from individuals with metabolic syndrome and chronic kidney disease. The enrichment of proximal tubule epithelial cells with phospholipids and multilamellar bodies was accompanied by enhanced inflammation, fibrosis, tubular damage markers, and higher urinary electrolyte content. Concomitantly to the intralysosomal lipid storage, a renal transcriptional response was initiated to enhance lysosomal degradation and lipid synthesis. In cultured proximal tubule epithelial cells, inhibition of cholesterol efflux transport or oxysterol treatment induced effects very similar to the in vivo situation, such as multilamellar body and phospholipid amassing, and induction of damage, inflammatory, fibrotic, and lipogenic molecules. The onset of phospholipidosis in proximal tubule epithelial cells is a novel pathological trait in metabolic syndrome-related chronic kidney disease, and emphasises the importance of healthy lysosomes and nutrition for kidney well-being. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Hypercholesterolemia/complications , Kidney Tubules, Proximal/metabolism , Lysosomes/metabolism , Obesity/complications , Phospholipids/adverse effects , Renal Insufficiency, Chronic/etiology , Animals , Case-Control Studies , Cell Line , Cholesterol, Dietary/metabolism , Disease Models, Animal , Fibrosis , Kidney Tubules, Proximal/ultrastructure , Lysosomes/ultrastructure , Male , Mice, Inbred C57BL , Mice, Transgenic , Phospholipids/metabolism , Proteomics/methods , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
13.
Kidney Int ; 92(4): 850-863, 2017 10.
Article En | MEDLINE | ID: mdl-28545716

Lymphangiogenesis is correlated with the degree of renal interstitial fibrosis. Pro-fibrotic transforming growth factor ß induces VEGF-C production, the main driver of lymphangiogenesis. Connective tissue growth factor (CTGF) is an important determinant of fibrotic tissue remodeling, but its possible involvement in lymphangiogenesis has not been explored. We found prominent lymphangiogenesis during tubulointerstitial fibrosis to be associated with increased expression of CTGF and VEGF-C in human obstructed nephropathy as well as in diabetic kidney disease. Using CTGF knockout mice, we investigated the involvement of CTGF in development of fibrosis and associated lymphangiogenesis in obstructive nephropathy. The increase of lymphatic vessels and VEGF-C in obstructed kidneys was significantly reduced in CTGF knockout compared to wild-type mice. Also in mouse kidneys subjected to ischemia-reperfusion injury, CTGF knockdown was associated with reduced lymphangiogenesis. In vitro, CTGF induced VEGF-C production in HK-2 cells, while CTGF siRNA suppressed transforming growth factor ß1-induced VEGF-C upregulation. Furthermore, surface plasmon resonance analysis showed that CTGF and VEGF-C directly interact. Interestingly, VEGF-C-induced capillary-like tube formation by human lymphatic endothelial cells was suppressed by full-length CTGF but not by naturally occurring proteolytic CTGF fragments. Thus, CTGF is significantly involved in fibrosis-associated renal lymphangiogenesis through regulation of, and direct interaction with, VEGF-C.


Connective Tissue Growth Factor/metabolism , Kidney Diseases/pathology , Kidney Tubules/pathology , Lymphangiogenesis , Vascular Endothelial Growth Factor C/metabolism , Animals , Cell Line , Connective Tissue Growth Factor/genetics , Disease Models, Animal , Fibrosis , Humans , Kidney Diseases/etiology , Kidney Diseases/surgery , Male , Mice , Mice, Knockout , Middle Aged , Protein Binding , RNA, Small Interfering/metabolism , Reperfusion Injury/complications , Signal Transduction , Surface Plasmon Resonance , Transforming Growth Factor beta1/metabolism , Up-Regulation
14.
Diabetes ; 65(9): 2591-605, 2016 09.
Article En | MEDLINE | ID: mdl-27325287

Aberrant mitochondrial fission plays a pivotal role in the pathogenesis of skeletal muscle insulin resistance. However, fusion-fission dynamics are physiologically regulated by inherent tissue-specific and nutrient-sensitive processes that may have distinct or even opposing effects with respect to insulin sensitivity. Based on a combination of mouse population genetics and functional in vitro assays, we describe here a regulatory circuit in which peroxisome proliferator-activated receptor γ (PPARγ), the adipocyte master regulator and receptor for the thiazolidinedione class of antidiabetic drugs, controls mitochondrial network fragmentation through transcriptional induction of Bnip3. Short hairpin RNA-mediated knockdown of Bnip3 in cultured adipocytes shifts the balance toward mitochondrial elongation, leading to compromised respiratory capacity, heightened fatty acid ß-oxidation-associated mitochondrial reactive oxygen species generation, insulin resistance, and reduced triacylglycerol storage. Notably, the selective fission/Drp1 inhibitor Mdivi-1 mimics the effects of Bnip3 knockdown on adipose mitochondrial bioenergetics and glucose disposal. We further show that Bnip3 is reciprocally regulated in white and brown fat depots of diet-induced obesity and leptin-deficient ob/ob mouse models. Finally, Bnip3(-/-) mice trade reduced adiposity for increased liver steatosis and develop aggravated systemic insulin resistance in response to high-fat feeding. Together, our data outline Bnip3 as a key effector of PPARγ-mediated adipose mitochondrial network fragmentation, improving insulin sensitivity and limiting oxidative stress.


Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , PPAR gamma/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Female , Glucose/metabolism , Immunoblotting , Immunohistochemistry , Insulin/metabolism , Insulin Resistance/genetics , Insulin Resistance/physiology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/genetics , Obesity/genetics , Obesity/metabolism , PPAR gamma/genetics , Radioimmunoprecipitation Assay , Reverse Transcriptase Polymerase Chain Reaction
15.
Perit Dial Int ; 36(6): 614-622, 2016.
Article En | MEDLINE | ID: mdl-27147290

♦ INTRODUCTION: Chronic uremia and the exposure to dialysis solutions during peritoneal dialysis (PD) induce peritoneal alterations. Using a long-term peritoneal exposure model, we compared the effects of chronic kidney failure (CKD) itself and exposure to either a 'conventional' or a 'biocompatible' dialysis solution on peritoneal morphology and function. ♦ METHODS: Wistar rats (Harlan, Zeist, the Netherlands) were grouped into: normal kidney function (NKF), CKD induced by 70% nephrectomy, CKD receiving daily peritoneal infusions with 3.86% glucose Dianeal (CKDD), or Physioneal (both solutions from Baxter Healthcare, Castlebar, Ireland) (CKDP). At 16 weeks, a peritoneal function test was performed, and histology, ultrastructure, and hydroxyproline content of peritoneal tissue were assessed. ♦ RESULTS: Comparing CKD with NKF, peritoneal transport rates were higher, mesothelial cells (MC) displayed increased number of microvilli, blood and lymph vasculature expanded, vascular basal lamina appeared thicker, with limited areas of duplication, and fibrosis had developed. All alterations, except lymphangiogenesis, were enhanced by exposure to both dialysis fluids. Distinct MC alterations were observed in CKDD and CKDP, the latter displaying prominent basolateral protrusions. In addition, CKDP was associated with a trend towards less fibrosis compared to CKDD. ♦ CONCLUSIONS: Chronic kidney failure itself induced peritoneal alterations, which were in part augmented by exposure to glucose-based dialysis solutions. Overall, the conventional and biocompatible solutions had similar long-term effects on the peritoneum. Importantly, the latter may attenuate the development of fibrosis.


Dialysis Solutions/pharmacology , Epithelium/pathology , Kidney Failure, Chronic/therapy , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/pathology , Analysis of Variance , Animals , Biopsy, Needle , Chi-Square Distribution , Dialysis Solutions/adverse effects , Disease Models, Animal , Epithelium/drug effects , Immunohistochemistry , Kidney Function Tests , Male , Nephrectomy/methods , Peritoneal Dialysis/methods , Peritoneal Fibrosis/etiology , Random Allocation , Rats , Rats, Wistar , Risk Factors , Statistics, Nonparametric
16.
PLoS One ; 11(1): e0147208, 2016.
Article En | MEDLINE | ID: mdl-26771826

Impaired function of NPC1 or NPC2 lysosomal proteins leads to the intracellular accumulation of unesterified cholesterol, the primary defect underlying Niemann-Pick type C (NPC) disease. In addition, glycosphingolipids (GSLs) accumulate in lysosomes as well. Intralysosomal lipid accumulation triggers the activation of a set of genes, including potential biomarkers. Transcript levels of Gpnmb have been shown to be elevated in various tissues of an NPC mouse model. We speculated that Gpnmb could serve as a marker for visceral lipid accumulation in NPC disease. We report that Gpnmb expression is increased at protein level in macrophages in the viscera of Npc1nih/nih mice. Interestingly, soluble Gpnmb was also found to be increased in murine and NPC patient plasma. Exposure of RAW264.7 macrophages to the NPC-phenotype-inducing drug U18666A also upregulated Gpnmb expression. Inhibition of GSL synthesis with the glucosylceramide synthase (GCS) inhibitor N-butyl-1-deoxynojirimycin prevented U18666A-induced Gpnmb induction and secretion. In summary, we show that Gpnmb is upregulated in NPC mice and patients, most likely due to GSL accumulation.


Biomarkers/metabolism , Eye Proteins/metabolism , Membrane Glycoproteins/metabolism , Niemann-Pick Disease, Type C/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line , Cholesterol/metabolism , Disease Models, Animal , Eye Proteins/genetics , Female , Foam Cells/metabolism , Glycosphingolipids/metabolism , Humans , Male , Membrane Glycoproteins/genetics , Mice , Middle Aged , Niemann-Pick Disease, Type C/genetics , Young Adult
17.
Adv Perit Dial ; 31: 21-5, 2015.
Article En | MEDLINE | ID: mdl-26714383

Increased lymphatic absorption might contribute to ultrafiltration failure in peritoneal dialysis (PD). Lymphangiogenesis develops during PD, but little is known about the relationship between its morphologic and functional parameters. The relationships between lymph vessel density, the effective lymphatic absorption rate (ELAR), and fibrosis were investigated in a rat model of chronic kidney failure (CKD) with exposure to dialysis solutions. Wistar rats (n = 44) were allocated to these groups: NKF (normal kidney function), CKD (70% nephrectomy), CKDD [CKD, with daily intraperitoneal (i.p.) Dianeal 3.86% (Baxter Healthcare BV, Utrecht, Netherlands)], CKDP [CKD, with daily i.p. Physioneal 3.86% (Baxter Healthcare BV)]. After 16 weeks, a peritoneal function test was performed, and the ELAR was calculated from the disappearance rate of i.p. dextran 70. The lymph vessel profile density (LVPD) was assessed using STEPanizer image analysis (Java application from Tschanz SA, Bern, Germany) of omental sections after anti-podoplanin immunostaining. Fibrosis was quantified by picro-sirius red staining. The LVPD was significantly increased in CKD rats compared with NKF rats, and no additional effect of dialysis solutions was present. The ELAR was increased in uremic rats compared with NKF rats. For all rats together, the LVPD correlated positively with the ELAR and with the amount of fibrosis. Chronic kidney disease itself induces lymphangiogenesis and fibrosis and increases the ELAR, independent of exposure to dialysis fluids. The ELAR is related to the LVPD in peritoneal tissue.


Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Lymphangiogenesis/physiology , Peritoneal Dialysis , Peritoneum/metabolism , Peritoneum/pathology , Animals , Dextrans/metabolism , Dialysis Solutions/metabolism , Disease Models, Animal , Fibrosis , Kidney Failure, Chronic/pathology , Male , Plasma Substitutes/metabolism , Rats , Rats, Wistar
18.
PLoS One ; 10(9): e0138107, 2015.
Article En | MEDLINE | ID: mdl-26418157

Gaucher disease is characterized by lysosomal accumulation of glucosylceramide due to deficient activity of lysosomal glucocerebrosidase (GBA). In cells, glucosylceramide is also degraded outside lysosomes by the enzyme glucosylceramidase 2 (GBA2) of which inherited deficiency is associated with ataxias. The interest in GBA and glucosylceramide metabolism in the brain has grown following the notion that mutations in the GBA gene impose a risk factor for motor disorders such as α-synucleinopathies. We earlier developed a ß-glucopyranosyl-configured cyclophellitol-epoxide type activity based probe (ABP) allowing in vivo and in vitro visualization of active molecules of GBA with high spatial resolution. Labeling occurs through covalent linkage of the ABP to the catalytic nucleophile residue in the enzyme pocket. Here, we describe a method to visualize active GBA molecules in rat brain slices using in vivo labeling. Brain areas related to motor control, like the basal ganglia and motor related structures in the brainstem, show a high content of active GBA. We also developed a ß-glucopyranosyl cyclophellitol-aziridine ABP allowing in situ labeling of GBA2. Labeled GBA2 in brain areas can be identified and quantified upon gel electrophoresis. The distribution of active GBA2 markedly differs from that of GBA, being highest in the cerebellar cortex. The histological findings with ABP labeling were confirmed by biochemical analysis of isolated brain areas. In conclusion, ABPs offer sensitive tools to visualize active GBA and to study the distribution of GBA2 in the brain and thus may find application to establish the role of these enzymes in neurodegenerative disease conditions such as α-synucleinopathies and cerebellar ataxia.


Brain/enzymology , Gaucher Disease/genetics , Glucosylceramidase/metabolism , Glucosylceramides/metabolism , Neurodegenerative Diseases/genetics , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Brain/metabolism , Cells, Cultured , Cerebellar Ataxia/genetics , Cerebellar Ataxia/pathology , Fluorescent Antibody Technique , Fluorescent Dyes/chemistry , Gaucher Disease/pathology , Glucosylceramidase/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/enzymology , Microglia/metabolism , Microscopy, Confocal , Neurodegenerative Diseases/pathology , Purkinje Cells/metabolism , Rats , Rats, Wistar
19.
PLoS One ; 10(8): e0135889, 2015.
Article En | MEDLINE | ID: mdl-26275242

The enzyme glucocerebrosidase (GBA) hydrolyses glucosylceramide (GlcCer) in lysosomes. Markedly reduced GBA activity is associated with severe manifestations of Gaucher disease including neurological involvement. Mutations in the GBA gene have recently also been identified as major genetic risk factor for Parkinsonism. Disturbed metabolism of GlcCer may therefore play a role in neuropathology. Besides lysosomal GBA, cells also contain a non-lysosomal glucosylceramidase (GBA2). Given that the two ß-glucosidases share substrates, we speculated that over-activity of GBA2 during severe GBA impairment might influence neuropathology. This hypothesis was studied in Niemann-Pick type C (Npc1-/-) mice showing secondary deficiency in GBA in various tissues. Here we report that GBA2 activity is indeed increased in the brain of Npc1-/- mice. We found that GBA2 is particularly abundant in Purkinje cells (PCs), one of the most affected neuronal populations in NPC disease. Inhibiting GBA2 in Npc1-/- mice with a brain-permeable low nanomolar inhibitor significantly improved motor coordination and extended lifespan in the absence of correction in cholesterol and ganglioside abnormalities. This trend was recapitulated, although not to full extent, by introducing a genetic loss of GBA2 in Npc1-/- mice. Our findings point to GBA2 activity as therapeutic target in NPC.


Glucosylceramides/metabolism , Niemann-Pick Disease, Type C/enzymology , beta-Glucosidase/metabolism , Animals , Glucosylceramides/genetics , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Proteins/genetics , Proteins/metabolism , Purkinje Cells/enzymology , Purkinje Cells/pathology , beta-Glucosidase/genetics
20.
J Diabetes Res ; 2015: 539787, 2015.
Article En | MEDLINE | ID: mdl-26171399

Connective tissue growth factor (CTGF; CCN2) plays a role in the development of diabetic nephropathy (DN). Urinary CTGF (uCTGF) is elevated in DN patients and has been proposed as a biomarker for disease progression, but it is unknown which pathophysiological factors contribute to elevated uCTGF. We studied renal handling of CTGF by infusion of recombinant CTGF in diabetic mice. In addition, uCTGF was measured in type 1 DN patients and compared with glomerular and tubular dysfunction and damage markers. In diabetic mice, uCTGF was increased and fractional excretion (FE) of recombinant CTGF was substantially elevated indicating reduced tubular reabsorption. FE of recombinant CTGF correlated with excretion of endogenous CTGF. CTGF mRNA was mainly localized in glomeruli and medullary tubules. Comparison of FE of endogenous and recombinant CTGF indicated that 60% of uCTGF had a direct renal source, while 40% originated from plasma CTGF. In DN patients, uCTGF was independently associated with markers of proximal and distal tubular dysfunction and damage. In conclusion, uCTGF in DN is elevated as a result of both increased local production and reduced reabsorption due to tubular dysfunction. We submit that uCTGF is a biomarker reflecting both glomerular and tubulointerstitial hallmarks of diabetic kidney disease.


Connective Tissue Growth Factor/urine , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/urine , Kidney Tubules, Distal/pathology , Kidney Tubules, Proximal/pathology , Up-Regulation , Adult , Animals , Biomarkers/urine , Cohort Studies , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Female , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Kidney Tubules, Distal/metabolism , Kidney Tubules, Distal/physiopathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/physiopathology , Male , Mice, Inbred C57BL , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/urine , Renal Elimination , Renal Reabsorption
...