Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Kidney Int ; 101(3): 574-584, 2022 03.
Article En | MEDLINE | ID: mdl-34767831

Sortilin, an intracellular sorting receptor, has been identified as a cardiovascular risk factor in the general population. Patients with chronic kidney disease (CKD) are highly susceptible to develop cardiovascular complications such as calcification. However, specific CKD-induced posttranslational protein modifications of sortilin and their link to cardiovascular calcification remain unknown. To investigate this, we examined two independent CKD cohorts for carbamylation of circulating sortilin and detected increased carbamylated sortilin lysine residues in the extracellular domain of sortilin with kidney function decline using targeted mass spectrometry. Structure analysis predicted altered ligand binding by carbamylated sortilin, which was verified by binding studies using surface plasmon resonance measurement, showing an increased affinity of interleukin 6 to in vitro carbamylated sortilin. Further, carbamylated sortilin increased vascular calcification in vitro and ex vivo that was accelerated by interleukin 6. Imaging by mass spectrometry of human calcified arteries revealed in situ carbamylated sortilin. In patients with CKD, sortilin carbamylation was associated with coronary artery calcification, independent of age and kidney function. Moreover, patients with carbamylated sortilin displayed significantly faster progression of coronary artery calcification than patients without sortilin carbamylation. Thus, carbamylated sortilin may be a risk factor for cardiovascular calcification and may contribute to elevated cardiovascular complications in patients with CKD.


Renal Insufficiency, Chronic , Vascular Calcification , Adaptor Proteins, Vesicular Transport , Humans , Protein Carbamylation , Protein Processing, Post-Translational , Vascular Calcification/etiology
2.
Cells ; 10(8)2021 08 16.
Article En | MEDLINE | ID: mdl-34440866

BACKGROUND: Vascular calcification is an active process that increases cardiovascular disease (CVD) risk. There is still no consensus on an appropriate biomarker for vascular calcification. We reasoned that the biomarker for vascular calcification is the collection of all blood components that can be sensed and integrated into a calcification response by human vascular smooth muscle cells (hVSMCs). METHODS: We developed a new cell-based high-content assay, the BioHybrid assay, to measure in vitro calcification. The BioHybrid assay was compared with the o-Cresolphthalein assay and the T50 assay. Serum and plasma were derived from different cohort studies including chronic kidney disease (CKD) stages III, IV, V and VD (on dialysis), pseudoxanthoma elasticum (PXE) and other cardiovascular diseases including serum from participants with mild and extensive coronary artery calcification (CAC). hVSMCs were exposed to serum and plasma samples, and in vitro calcification was measured using AlexaFluor®-546 tagged fetuin-A as calcification sensor. RESULTS: The BioHybrid assay measured the kinetics of calcification in contrast to the endpoint o-Cresolphthalein assay. The BioHybrid assay was more sensitive to pick up differences in calcification propensity than the T50 assay as determined by measuring control as well as pre- and post-dialysis serum samples of CKD patients. The BioHybrid response increased with CKD severity. Further, the BioHybrid assay discriminated between calcification propensity of individuals with a high CAC index and individuals with a low CAC index. Patients with PXE had an increased calcification response in the BioHybrid assay as compared to both spouse and control plasma samples. Finally, vitamin K1 supplementation showed lower in vitro calcification, reflecting changes in delta Agatston scores. Lower progression within the BioHybrid and on Agatston scores was accompanied by lower dephosphorylated-uncarboxylated matrix Gla protein levels. CONCLUSION: The BioHybrid assay is a novel approach to determine the vascular calcification propensity of an individual and thus may add to personalised risk assessment for CVD.


Muscle, Smooth, Vascular/metabolism , Vascular Calcification/blood , Biomarkers/blood , Calcium-Binding Proteins/blood , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/drug therapy , Cells, Cultured , Extracellular Matrix Proteins/blood , Fluorescent Dyes/chemistry , Hematologic Tests , Humans , Kinetics , Renal Dialysis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/therapy , Vascular Calcification/diagnosis , Vitamin K 1/therapeutic use , alpha-2-HS-Glycoprotein/chemistry , alpha-2-HS-Glycoprotein/metabolism , Matrix Gla Protein
3.
J Clin Med ; 10(14)2021 Jul 09.
Article En | MEDLINE | ID: mdl-34300217

In COVID-19, guidelines recommend a urinalysis on hospital admission as SARS-CoV-2 renal tropism, post-mortem, was associated with disease severity and mortality. Following the hypothesis from our pilot study, we now validate an algorithm harnessing urinalysis to predict the outcome and the need for ICU resources on admission to hospital. Patients were screened for urinalysis, serum albumin (SA) and antithrombin III activity (AT-III) obtained prospectively on admission. The risk for an unfavorable course was categorized as (1) "low", (2) "intermediate" or (3) "high", depending on (1) normal urinalysis, (2) abnormal urinalysis with SA ≥ 2 g/dL and AT-III ≥ 70%, or (3) abnormal urinalysis with SA or AT-III abnormality. Time to ICU admission or death served as the primary endpoint. Among 223 screened patients, 145 were eligible for enrollment, 43 falling into the low, 84 intermediate, and 18 into high-risk categories. An abnormal urinalysis significantly elevated the risk for ICU admission or death (63.7% vs. 27.9%; HR 2.6; 95%-CI 1.4 to 4.9; p = 0.0020) and was 100% in the high-risk group. Having an abnormal urinalysis was associated with mortality, a need for mechanical ventilation, extra-corporeal membrane oxygenation or renal replacement therapy. In conclusion, our data confirm that COVID-19-associated urine abnormalities on admission predict disease aggravation and the need for ICU (ClinicalTrials.gov number NCT04347824).

4.
PLoS One ; 15(2): e0228938, 2020.
Article En | MEDLINE | ID: mdl-32074140

Calcifications can disrupt organ function in the cardiovascular system and the kidney, and are particularly common in patients with chronic kidney disease (CKD). Fetuin-A deficient mice maintained against the genetic background DBA/2 exhibit particularly severe soft tissue calcifications, while fetuin-A deficient C57BL/6 mice remain healthy. We employed molecular genetic analysis to identify risk factors of calcification in fetuin-A deficient mice. We sought to identify pharmaceutical therapeutic targets that could be influenced by dietary of parenteral supplementation. We studied the progeny of an intercross of fetuin-A deficient DBA/2 and C57BL/6 mice to identify candidate risk genes involved in calcification. We determined that a hypomorphic mutation of the Abcc6 gene, a liver ATP transporter supplying systemic pyrophosphate, and failure to regulate the Trpm6 magnesium transporter in kidney were associated with severity of calcification. Calcification prone fetuin-A deficient mice were alternatively treated with parenteral administration of fetuin-A dietary magnesium supplementation, phosphate restriction, or by or parenteral pyrophosphate. All treatments markedly reduced soft tissue calcification, demonstrated by computed tomography, histology and tissue calcium measurement. We show that pathological ectopic calcification in fetuin-A deficient DBA/2 mice is caused by a compound deficiency of three major extracellular and systemic inhibitors of calcification, namely fetuin-A, magnesium, and pyrophosphate. All three of these are individually known to contribute to stabilize protein-mineral complexes and thus inhibit mineral precipitation from extracellular fluid. We show for the first time a compound triple deficiency that can be treated by simple dietary or parenteral supplementation. This is of special importance in patients with advanced CKD, who commonly exhibit reduced serum fetuin-A, magnesium and pyrophosphate levels.


Calcinosis/pathology , Microvessels/physiology , alpha-2-HS-Glycoprotein/metabolism , Animals , Calcinosis/genetics , Diphosphates/metabolism , Disease Models, Animal , Female , Kidney/pathology , Liver/pathology , Magnesium/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microvessels/metabolism , Minerals , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Renal Insufficiency, Chronic/complications , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , alpha-2-HS-Glycoprotein/physiology , alpha-Fetoproteins
5.
Hypertension ; 70(2): 412-419, 2017 08.
Article En | MEDLINE | ID: mdl-28652472

Despite advancements in lowering blood pressure, the best approach to lower it remains controversial because of the lack of information on the molecular basis of hypertension. We, therefore, performed plasma proteomics of plasma from patients with hypertension to identify molecular determinants detectable in these subjects but not in controls and vice versa. Plasma samples from hypertensive subjects (cases; n=118) and controls (n=85) from the InGenious HyperCare cohort were used for this study and performed mass spectrometric analysis. Using biostatistical methods, plasma peptides specific for hypertension were identified, and a model was developed using least absolute shrinkage and selection operator logistic regression. The underlying peptides were identified and sequenced off-line using matrix-assisted laser desorption ionization orbitrap mass spectrometry. By comparison of the molecular composition of the plasma samples, 27 molecular determinants were identified differently expressed in cases from controls. Seventy percent of the molecular determinants selected were found to occur less likely in hypertensive patients. In cross-validation, the overall R2 was 0.434, and the area under the curve was 0.891 with 95% confidence interval 0.8482 to 0.9349, P<0.0001. The mean values of the cross-validated proteomic score of normotensive and hypertensive patients were found to be -2.007±0.3568 and 3.383±0.2643, respectively, P<0.0001. The molecular determinants were successfully identified, and the proteomic model developed shows an excellent discriminatory ability between hypertensives and normotensives. The identified molecular determinants may be the starting point for further studies to clarify the molecular causes of hypertension.


Biostatistics/methods , Blood Pressure/physiology , Hypertension , Proteomics/methods , Adult , Antihypertensive Agents/therapeutic use , Case-Control Studies , Confidence Intervals , Europe , Female , Humans , Hypertension/diagnosis , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/physiopathology , Male , Middle Aged , Models, Biological , Molecular Diagnostic Techniques , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/statistics & numerical data
6.
Clin J Am Soc Nephrol ; 12(2): 315-322, 2017 02 07.
Article En | MEDLINE | ID: mdl-27940458

BACKGROUND AND OBJECTIVES: Patients receiving hemodialysis are at risk of cardiovascular events. A novel blood test (T50 test) determines the individual calcification propensity of blood. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: T50 was determined in 2785 baseline serum samples of patients receiving hemodialysis enrolled in the Evaluation of Cinacalcet Therapy to Lower Cardiovascular Events (EVOLVE) trial and the T50 results were related to patient outcomes. RESULTS: Serum albumin, bicarbonate, HDL cholesterol, and creatinine were the main factors positively/directly and phosphate was the main factor negatively/inversely associated with T50. The primary composite end point (all-cause mortality, myocardial infarction [MI], hospitalization for unstable angina, heart failure, or peripheral vascular event [PVE]) was reached in 1350 patients after a median follow-up time of 619 days. After adjustments for confounding, a lower T50 was independently associated with a higher risk of the primary composite end point as a continuous measure (hazard ratio [HR] per 1 SD lower T50, 1.15; 95% confidence interval [95% CI], 1.08 to 1.22; P<0.001). Furthermore, lower T50 was associated with a higher risk in all-cause mortality (HR per 1 SD lower T50, 1.10; 95% CI, 1.02 to 1.17; P=0.001), MI (HR per 1 SD lower T50, 1.38; 95% CI, 1.19 to 1.60; P<0.001), and PVE (HR per 1 SD lower T50, 1.22; 95% CI, 1.05 to 1.42; P=0.01). T50 improved risk prediction (integrated discrimination improvement and net reclassification improvement, P<0.001 and P=0.001) of the primary composite end point. CONCLUSIONS: Blood calcification propensity was independently associated with the primary composite end point, all-cause mortality, MI, and PVE in the EVOLVE study and improved risk prediction. Prospective trials should clarify whether T50-guided therapies improve outcomes.


Calcinosis/blood , Cardiovascular Diseases/blood , Cardiovascular Diseases/epidemiology , Cause of Death , Renal Dialysis , Adult , Aged , Angina, Unstable/blood , Angina, Unstable/epidemiology , Calcimimetic Agents/therapeutic use , Cardiovascular Diseases/therapy , Cinacalcet/therapeutic use , Female , Heart Failure/blood , Heart Failure/epidemiology , Hematologic Tests , Hospitalization/statistics & numerical data , Humans , Kidney Failure, Chronic/therapy , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/epidemiology , Peripheral Vascular Diseases/blood , Peripheral Vascular Diseases/epidemiology , Peripheral Vascular Diseases/surgery , Predictive Value of Tests , Prospective Studies , Survival Rate
7.
J Am Soc Nephrol ; 27(6): 1650-64, 2016 06.
Article En | MEDLINE | ID: mdl-26453615

Pathologic proliferation of mesangial and parietal epithelial cells (PECs) is a hallmark of various glomerulonephritides. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that mediates inflammation by engagement of a receptor complex involving the components CD74, CD44, CXCR2, and CXCR4. The proliferative effects of MIF may involve CD74 together with the coreceptor and PEC activation marker CD44. Herein, we analyzed the effects of local glomerular MIF/CD74/CD44 signaling in proliferative glomerulonephritides. MIF, CD74, and CD44 were upregulated in the glomeruli of patients and mice with proliferative glomerulonephritides. During disease, CD74 and CD44 were expressed de novo in PECs and colocalized in both PECs and mesangial cells. Stress stimuli induced MIF secretion from glomerular cells in vitro and in vivo, in particular from podocytes, and MIF stimulation induced proliferation of PECs and mesangial cells via CD74. In murine crescentic GN, Mif-deficient mice were almost completely protected from glomerular injury, the development of cellular crescents, and the activation and proliferation of PECs and mesangial cells, whereas wild-type mice were not. Bone marrow reconstitution studies showed that deficiency of both nonmyeloid and bone marrow-derived Mif reduced glomerular cell proliferation and injury. In contrast to wild-type mice, Cd74-deficient mice also were protected from glomerular injury and ensuing activation and proliferation of PECs and mesangial cells. Our data suggest a novel molecular mechanism and glomerular cell crosstalk by which local upregulation of MIF and its receptor complex CD74/CD44 mediate glomerular injury and pathologic proliferation in GN.


Antigens, Differentiation, B-Lymphocyte/physiology , Glomerulonephritis/etiology , Histocompatibility Antigens Class II/physiology , Macrophage Migration-Inhibitory Factors/physiology , Animals , Cell Proliferation , Cells, Cultured , Female , Glomerulonephritis/pathology , Kidney Glomerulus/cytology , Male , Mice , Mice, Inbred C57BL
8.
Nephron ; 129(4): 247-52, 2015.
Article En | MEDLINE | ID: mdl-25823466

BACKGROUND/AIMS: Vascular calcifications are frequently found among dialysis patients, and the calcification process may influence the patient's outcome. The aim of the present study was to determine the role that vascular calcifications may have on autologous arteriovenous fistula (AVF) survival. METHODS: This study included 90 patients (49 males, mean age 62 ± 11) with a native AVF treated by chronic hemodialysis (HD) for more than one year. The overall vascular calcification scores ranged from 0-11 (Adragao score + vascular access calcification score); patients were categorized into mild (score 0-3; n = 36), moderate (score 4-7; n = 24) and severe (score 8-11; n = 30) calcification groups. AVF survival was then followed for 5 years after calcification measurement or until the patient's death/transplantation. RESULTS: Patients with more pronounced vascular calcifications were more frequently diabetic and male. Multiple linear regression analysis showed a significant relationship between calcification score and male gender, diabetes mellitus, previous duration of AVF, low dialysis flow rate and intact parathormone (iPTH) values. After multivariate adjustment for basal differences, Cox proportional analysis revealed a graded impact of calcification scores on AVF failure: moderate scores (were associated with a hazard rate (HR) of 3.82 (95% confidence interval (CI) 1.10-13.23) and severe scores with an HR of 4.65 (CI 0.97-22.38). CONCLUSION: Vascular calcifications are associated with worse survival of native arteriovenous hemodialysis fistulas.


Arteriovenous Fistula/mortality , Renal Dialysis , Vascular Calcification/etiology , Aged , Female , Follow-Up Studies , Humans , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/mortality , Kidney Failure, Chronic/therapy , Male , Middle Aged , Pulse Wave Analysis , Renal Dialysis/adverse effects , Vascular Calcification/pathology
9.
PLoS One ; 9(3): e92115, 2014.
Article En | MEDLINE | ID: mdl-24667162

Mesenchymal stem cell (MSC) transplantation has the potential for organ repair. Nevertheless, some factors might lessen the regenerative potential of MSCs, e.g. donor age or systemic disease. It is thus important to carefully assess the patient's suitability for autologous MSC transplantation. Here we investigated the effects of chronic kidney disease (CKD) on MSC function. We isolated bone marrow MSCs from remnant kidney rats (RK) with CKD (CKD-RK-MSC) and found signs of premature senescence: spontaneous adipogenesis, reduced proliferation capacity, active senescence-associated-ß-galactosidase, accumulation of actin and a modulated secretion profile. The functionality of CKD-RK-MSCs in vivo was tested in rats with acute anti-Thy1.1-nephritis, where healthy MSCs have been shown to be beneficial. Rats received healthy MSCs, CKD-RK-MSC or medium by injection into the left renal artery. Kidneys receiving healthy MSCs exhibited accelerated healing of glomerular lesions, whereas CKD-RK-MSC or medium exerted no benefit. The negative influence of advanced CKD/uremia on MSCs was confirmed in a second model of CKD, adenine nephropathy (AD). MSCs from rats with adenine nephropathy (CKD-AD-MSC) also exhibited cellular modifications and functional deficits in vivo. We conclude that CKD leads to a sustained loss of in vitro and in vivo functionality in MSCs, possibly due to premature cellular senescence. Considering autologous MSC therapy in human renal disease, studies identifying uremia-associated mechanisms that account for altered MSC function are urgently needed.


Cellular Senescence/physiology , Kidney Glomerulus/pathology , Mesenchymal Stem Cells/cytology , Nephritis/pathology , Regeneration/physiology , Renal Insufficiency, Chronic/pathology , Animals , Blotting, Western , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Fibroblasts/cytology , Humans , Immunoenzyme Techniques , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mesenchymal Stem Cell Transplantation , Nephritis/immunology , Nephritis/therapy , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , Renal Insufficiency, Chronic/therapy , Reverse Transcriptase Polymerase Chain Reaction , Thy-1 Antigens/immunology , Tissue Donors
10.
J Heart Valve Dis ; 22(3): 391-9, 2013 May.
Article En | MEDLINE | ID: mdl-24151766

BACKGROUND AND AIM OF THE STUDY: Cardiac valve calcification (CVC) and coronary artery calcification (CAC) appear to be linked pathogenetically, and both are associated with a poor prognosis among patients with chronic kidney disease on dialysis (CKD-5D). Little is known, however, about factors that affect the progression of CVC and CAC. METHODS: A post-hoc analysis was performed of the ADVANCE study to assess whether patients with CVC are more prone to CAC progression, and whether CVC predicts the response to different treatments for secondary hyperparathyroidism. RESULTS: Subjects were randomized to treatment with either cinacalcet and low doses of vitamin D analogs or larger, varying doses of vitamin D. Among 235 subjects, aortic valve or mitral valve calcification was detected in 108 (46%) and 118 (50%), respectively; 69 subjects (29%) had calcification of both valves. CVC was associated both with baseline CAC and CAC progression (p < 0.05). Subjects with aortic valve calcification who were treated with cinacalcet and low doses of vitamin D experienced less progression of CAC than subjects given larger, varying doses of vitamin D (adjusted OR: 0.26; 95% CI: 0.10, 0.64). This effect was greater in subjects with larger CAC burden at baseline. CONCLUSION: The study findings suggest that CVC is a predictor of CAC progression and, potentially, of greater cardiovascular vulnerability. Treatment with cinacalcet combined with low doses of vitamin D slowed the progression of CAC compared to therapy using larger, varying doses of vitamin D.


Calcinosis , Coronary Vessels/pathology , Heart Valves/pathology , Hyperparathyroidism, Secondary/drug therapy , Kidney Failure, Chronic/therapy , Naphthalenes , Renal Dialysis/adverse effects , Vitamin D , Aged , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/pharmacokinetics , Calcinosis/diagnosis , Calcinosis/epidemiology , Calcinosis/etiology , Calcinosis/prevention & control , Cinacalcet , Dose-Response Relationship, Drug , Drug Monitoring , Female , Humans , Hyperparathyroidism, Secondary/etiology , Hyperparathyroidism, Secondary/metabolism , Logistic Models , Male , Middle Aged , Naphthalenes/administration & dosage , Naphthalenes/pharmacokinetics , Prognosis , Receptors, Calcium-Sensing/agonists , Treatment Outcome , Vitamin D/administration & dosage , Vitamin D/pharmacokinetics
12.
Gut ; 61(8): 1209-18, 2012 Aug.
Article En | MEDLINE | ID: mdl-22287599

BACKGROUND: At present hepatocyte transplantation is a promising option for cellular therapy of end-stage liver diseases. However, the underlying molecular mechanisms need to be better defined in order to translate this technique into clinical use. This study investigated the cursiv relevance of hepatocyte growth factor (HGF)/c-Met signalling for hepatocyte repopulation after transplantion. METHODS: Wild-type mice (c-Met(loxP/loxP)) and hepatocyte-specific conditional c-Met (HGF receptor) knockout (c-Met(Δhepa)) mice were used as donors and recipients for hepatocyte transplantation. RESULTS: Transplantation experiments revealed two major findings. First it was demonstrated that c-Met is indispensable in donor cells, as c-Met(Δhepa) cells did not repopulate recipient livers after transplantation. Second, genetic deletion of c-Met in recipient hepatocytes resulted in enhanced expansion of unmodified donor cells in host livers (up to 250-fold after 12 weeks). The relevant mechanisms for this observation in c-Met(Δhepa) host hepatocytes could be defined. c-Met(Δhepa) hepatocytes showed enhanced apoptosis, reduced cellular proliferation and a lack of AKT-kinase and STAT3 activation. In addition, tissue remodelling was changed in c-Met(Δhepa) recipient livers. Therefore, the lack of pro-proliferative transcription factors, increased apoptosis and changes in matrix-remodelling inhibit host cell proliferation in c-Met(Δhepa) recipient livers and thus favour repopulation of transplanted hepatocytes. Therapeutically liver repopulation could be increased through adenoviral expression of NK-4--an inhibitor of HGF signalling--in host hepatocytes. CONCLUSION: HGF/c-Met plays a crucial role in host and donor cells of the liver for the cursiv selection of transplanted hepatocytes. Modulating HGF-dependent signalling seems a promising therapeutic option to favour expansion of transplanted hepatocytes.


Gene Expression Regulation , Hepatocyte Growth Factor/genetics , Hepatocytes/transplantation , Liver Regeneration , Liver Transplantation/methods , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Apoptosis , Blotting, Western , Cell Communication , Cell Proliferation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Hepatocyte Growth Factor/biosynthesis , Hepatocytes/cytology , In Situ Nick-End Labeling , Liver Failure/genetics , Liver Failure/metabolism , Liver Failure/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/biosynthesis , Signal Transduction
13.
Invest Ophthalmol Vis Sci ; 51(8): 4273-81, 2010 Aug.
Article En | MEDLINE | ID: mdl-20237249

PURPOSE: A number of key ocular diseases, including diabetic retinopathy and age-related macular degeneration, are characterized by localized areas of epithelial or endothelial damage, which can ultimately result in the growth of fragile new blood vessels, vitreous hemorrhage, and retinal detachment. VEGF-A(165), the principal neovascular agent in ocular angiogenic conditions, is formed by proximal splice site selection in its terminal exon 8. Alternative splicing of this exon results in an antiangiogenic isoform, VEGF-A(165)b, which is downregulated in diabetic retinopathy. Here the authors investigate the antiangiogenic activity of VEGF(165)b and its effect on retinal epithelial and endothelial cell survival. METHODS: VEGF-A(165)b was injected intraocularly in a mouse model of retinal neovascularization (oxygen-induced retinopathy [OIR]). Cytotoxicity and cell migration assays were used to determine the effect of VEGF-A(165)b. RESULTS: VEGF-A(165)b dose dependently inhibited angiogenesis (IC(50), 12.6 pg/eye) and retinal endothelial migration induced by 1 nM VEGF-A(165) across monolayers in culture (IC(50), 1 nM). However, it also acts as a survival factor for endothelial cells and retinal epithelial cells through VEGFR2 and can stimulate downstream signaling. Furthermore, VEGF-A(165)b injection, while inhibiting neovascular proliferation in the eye, reduced the ischemic insult in OIR (IC(50), 2.6 pg/eye). Unlike bevacizumab, pegaptanib did not interact directly with VEGF-A(165)b. CONCLUSIONS: The survival effects of VEGF-A(165)b signaling can protect the retina from ischemic damage. These results suggest that VEGF-A(165)b may be a useful therapeutic agent in ischemia-induced angiogenesis and a cytoprotective agent for retinal pigment epithelial cells.


Angiogenesis Inhibitors/pharmacology , Cell Movement/drug effects , Disease Models, Animal , Endothelium, Vascular/drug effects , Retinal Neovascularization/prevention & control , Vascular Endothelial Growth Factor A/pharmacology , Animals , Animals, Newborn , Aptamers, Nucleotide/therapeutic use , Cell Survival/drug effects , Cells, Cultured , Cytoprotection , Dose-Response Relationship, Drug , Drug Interactions , Epithelial Cells/drug effects , Half-Life , Humans , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , Rats , Recombinant Proteins/pharmacology , Retinal Neovascularization/metabolism , Retinal Vessels/cytology , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Nephrol Dial Transplant ; 25(1): 283-92, 2010 Jan.
Article En | MEDLINE | ID: mdl-19773417

BACKGROUND: Early and long-term use of cyclosporine A (CsA) leads to increased risks of renal toxicity. We hypothesized that administration of daclizumab in combination with mycophenolate mofetil (MMF) allows a relevant reduction in the dose of CsA. METHODS: We carried out a 3-year, prospective, randomized, controlled clinical multi-centre trial in 156 patients. The patients were randomized to standard treatment (CsA, MMF, steroids) or to high-dose daclizumab (first dose: 2 mg/kg), in combination with low-dose CsA, MMF and steroids. We maintained the mean CsA levels of daclizumab patients at 57% of standard patients (132 versus 216 ng/ml) on Day 7 post-transplant, and 84% by 6 months. RESULTS: Primary outcome, creatinine clearance (with imputation of informative dropouts) at 12 months, was significantly better in daclizumab-treated (34 +/- 17) than standard patients (29 +/- 17; P = 0.028, two sided). Only 5 cases of BPAR were recorded in the daclizumab compared to 22 in the standard group (P = 0.0016). Daclizumab patients had 91% event-free survival after 1 year compared to 66% in standard patients (P = 0.00017). CONCLUSION: We demonstrate here that high-dose daclizumab in combination with lower CsA levels in adult renal transplant recipients is as or more effective than standard regimen (CsA, MMF, steroids) and may result in better outcomes at 12 months post-transplant with no increase in adverse reactions.


Antibodies, Monoclonal/pharmacology , Cyclosporine/pharmacology , Immunoglobulin G/pharmacology , Kidney Transplantation , Kidney/drug effects , Kidney/physiology , Mycophenolic Acid/analogs & derivatives , Steroids/pharmacology , Adult , Aged , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Biopsy , Cyclosporine/therapeutic use , Daclizumab , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Graft Rejection/epidemiology , Graft Rejection/prevention & control , Humans , Immunoglobulin G/therapeutic use , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Incidence , Kidney/surgery , Kidney Transplantation/pathology , Longitudinal Studies , Male , Middle Aged , Mycophenolic Acid/pharmacology , Mycophenolic Acid/therapeutic use , Prospective Studies , Steroids/therapeutic use , Treatment Outcome
16.
J Am Soc Nephrol ; 16(10): 2985-96, 2005 Oct.
Article En | MEDLINE | ID: mdl-16093451

The pivotal role of PDGF-B for mesangioproliferative glomerular disease is well established. Here, Y-box protein-1 (YB-1) was identified as a downstream signaling target of PDGF-B. In healthy kidney cells, YB-1 was located predominantly within the nuclear compartment. Subsequent to PDGF-B infusion and in the course of anti-Thy1.1-induced mesangioproliferative glomerulonephritis, relocalization of YB-1 into the cytoplasm was observed. In experimental models that lack profound mesangial cell proliferation (e.g., Puromycin-nephrosis, passive Heyman nephritis, spontaneous normotensive nephrosclerosis, hyperlipidemic diabetic nephropathy), YB-1 remained nuclear. This translocation coincided with upregulation of YB-1 protein levels within the mesangial compartment. Increased YB-1 expression and subcellular shuttling was dependent on PDGF-B signaling via the mitogen-activated protein kinase pathway because these alterations were prevented by specific PDGF aptamers and the mitogen-activated protein kinase pathway inhibitor U0126. Furthermore, PDGF-B strongly induced YB-1 expression in vitro. This induction was important because RNAi-dependent knockdown of YB-1 abolished the mitogenic PDGF-B effect. Taken together, YB-1 seems to represent a specific and necessary PDGF-B target in mesangioproliferative glomerular disease.


Glomerular Mesangium , Glomerulonephritis, Membranoproliferative/etiology , Proto-Oncogene Proteins c-sis/physiology , Y-Box-Binding Protein 1/physiology , Animals , Cell Division , Cells, Cultured , Humans , Male , Mesangial Cells/cytology , Mesangial Cells/metabolism , Protein Transport , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Y-Box-Binding Protein 1/biosynthesis , Y-Box-Binding Protein 1/genetics
17.
J Am Soc Nephrol ; 16(6): 1733-41, 2005 Jun.
Article En | MEDLINE | ID: mdl-15829708

Podocyte loss contributes to the development of glomerulosclerosis. Although podocyte detachment has been recognized as a new mechanism of podocyte loss in glomerular diseases, its time course and relationship to disease activity are not known. Urinary excretion of viable podocytes was quantified in two models of transient glomerular injury, i.e., rats with puromycin aminonucleoside-induced nephrosis (PAN) and mesangioproliferative nephropathy (anti-Thy 1.1 nephritis model), as well as in a model of continuous glomerular injury, i.e., hypertensive nephropathy (5/6-nephrectomy model), and in aging rats. The number of glomerular Wilm's tumor (WT)-1-positive podocytes and the glomerular expression of cell-cycle proteins in vivo were assessed. Urinary podocyte loss occurred in both primary (PAN) and secondary (anti-Thy 1.1 nephritis) in parallel to the onset of proteinuria. However, subsequently proteinuria persisted despite remission of podocyturia. In continuous glomerular injury, i.e., after 5/6-nephrectomy, podocyturia paralleled the course of proteinuria and of systemic hypertension, whereas no podocyturia became detectable during normal aging (up to 12 mo). Despite podocyte detachment of varying degrees, no decrease in glomerular podocyte counts (i.e., WT-1 positive nuclei) was noted in either disease model. Podocyturia in the PAN and anti-Thy 1.1 nephritis model was preceded by entry of glomerular podocytes into the cell cycle, i.e., cyclin D1, cdc2, and/or proliferating cell nuclear antigen (PCNA) expression. Podocyturia is a widespread phenomenon in glomerular disease and not simply a reflection of proteinuria because it is limited to phases of ongoing glomerular injury. The data suggest that podocyturia may become a more sensitive means to assess the activity of glomerular damage than proteinuria.


Glomerulonephritis/physiopathology , Glomerulonephritis/urine , Urine/cytology , Aging/physiology , Animals , Epithelial Cells , Glomerulonephritis/complications , Kidney Glomerulus , Male , Models, Animal , Proteinuria/etiology , Proteinuria/physiopathology , Rats , Urine/chemistry
18.
Transplantation ; 77(10): 1566-71, 2004 May 27.
Article En | MEDLINE | ID: mdl-15239623

BACKGROUND: Bone mineral density (BMD) decreases significantly early after renal transplantation. This prospective study was designed to evaluate the long-term lumbar BMD development. METHODS: Sixty-three renal-transplant recipients (mean age 44 +/- 12 years, 37 [59%] male) underwent serial yearly posttransplant laboratory parameter and BMD measurements of the lumbar spine (dual energy x-ray absorptiometry). Combined maintenance immunosuppression included prednisolone in 95% of patients. The minimum number of consecutive scans was three; the maximum number seven (n = 15). Examinations were performed between 3 +/- 2 and 68 +/- 4 months posttransplant. RESULTS: BMD was significantly lower compared with healthy controls at all times after transplantation. t scores were below -1. BMD development revealed a biphasic pattern: between 3 +/- 2 and 10 +/- 2 months, a significant BMD decrease of -0.016 +/- 0.055 g/cm2 (-1.6%, P = 0.024) occurred. Later, a moderate increase resulting in BMD stability until the sixth year posttransplant was detected. Within the first year, posttransplant osteocalcin (from 19 +/- 15 to 32 +/- 23 microg/L) and calcitriol (from 24 +/- 15 to 43 +/- 24 ng/L) displayed a significant increase. Compared with patients with a pronounced decrease, patients with a substantial increase in early posttransplant BMD had a lower baseline BMD (0.989 +/- 0.131 vs. 1.149 +/- 0.202 g/cm2 [P = 0.0122]) and lower creatinine levels (105 +/- 23 vs. 141 +/- 53 mmol/L [P = 0.0227]). CONCLUSION: Our study confirms a significant decrease of lumbar BMD early after renal transplantation. Bone loss was less pronounced than previously described. The longitudinal follow-up verifies a previously assumed biphasic lumbar BMD development: after the first year, no further significant bone loss occurred, and bone density remained relatively stable at significantly lower levels compared with healthy controls.


Bone Density , Kidney Transplantation , Lumbar Vertebrae/metabolism , Absorptiometry, Photon , Adult , Calcitriol/blood , Creatinine/blood , Female , Follow-Up Studies , Humans , Male , Middle Aged , Osteocalcin/blood , Postoperative Period , Time Factors
19.
Am J Kidney Dis ; 40(5): 1066-74, 2002 Nov.
Article En | MEDLINE | ID: mdl-12407653

BACKGROUND: Rapid bone loss is a frequent finding early after kidney transplantation. Only limited data are available on the bone mineral density (BMD) in long-term kidney transplant recipients. METHODS: In 26 kidney transplant recipients (13 men and 13 women, age 45.3 +/- 12.3 years), serum biochemical markers of bone metabolism and BMD at the lumbar vertebrae L2-4 were evaluated prospectively in three serial examinations (E1, E2, E3; method: dual-energy X-ray absorptiometry). Examinations were performed at 47 +/- 2 months, 59 +/- 2 months, and 71 +/- 2 months after transplantation. All patients received standard dual or triple immunosuppression including prednisolone. RESULTS: The mean BMD was significantly lower (P < 0.001) than in sex-matched young controls: T-score was -1.43 +/- 1.49 (E1), -1.39 +/- 1.40 (E2), and -1.44 +/- 1.30 (E3). The BMD did not change significantly (Delta BMD, -0.5 +/- 5.9%) from E1 to E3. Regression analysis did not show significant associations between Delta BMD and biochemical parameters or prednisolone dosage. No clinically apparent new lumbar vertebral fracture occurred. The mean intact parathyroid hormone was 110.1 +/- 97.5 pg/mL (E1), 121 +/- 102.7 pg/mL (E2), and 134.5 +/- 128.6 pg/mL (E3). Serum creatinine was 1.44 +/- 0.45 (128 +/- 40) mg/dL (micromol/L) (E1), 1.44 +/- 0.47 (127 +/- 42) mg/dL (micromol/L) (E2), and 1.45 +/- 0.70 (128 +/- 62) mg/dL (micromol/L) (E3). Ten patients (38.5%) showed an increase of BMD (+5.7 +/- 3.2%) and 15 patients (57.7%) showed a decrease of -4.7 +/- 3.2% (P < 0.0001). Both groups were different in T-scores at E1 (-2.29 +/- 1 versus -0.88 +/- 1.5); intact parathyroid hormone, creatinine, vitamin D levels, and prednisolone dosage were not significantly different. CONCLUSION: This study shows that lumbar BMD is reduced in long-term kidney transplant recipients. During our 24-month observation period, overall lumbar BMD remained stable.


Bone Density/physiology , Kidney Transplantation , Kidney Transplantation/adverse effects , Lumbar Vertebrae/pathology , Absorptiometry, Photon/methods , Adult , Aged , Bone Resorption/blood , Bone Resorption/pathology , Bone and Bones/metabolism , Creatinine/blood , Creatinine/metabolism , Female , Humans , Kidney/physiopathology , Kidney Function Tests , Kidney Transplantation/methods , Lumbar Vertebrae/diagnostic imaging , Male , Middle Aged , Parathyroid Hormone/blood , Prospective Studies , Regression Analysis , Sex Factors , Time Factors
20.
Biochem J ; 362(Pt 3): 693-700, 2002 Mar 15.
Article En | MEDLINE | ID: mdl-11879197

Glomerular hypertension has been established as a major factor contributing to glomerular scarring. Underlying cellular mechanisms leading to matrix accumulation are largely unknown. The isolated effect of oscillating hyperbaric pressure [OP; P(max) 50 mmHg (1 mmHg=0.133 kPa), P(mean) 24 mmHg, with a fixed oscillation of 60/min] on matrix-degrading protease secretion by rat mesangial cells (MCs) was analysed using a pressure chamber model described previously [Mertens, Espenkott, Venjakob, Heintz, Handt and Sieberth (1998) Hypertension 32, 945-952]. MCs were grown under atmospheric pressure (AP) or a controlled OP, and protease synthesis and gene transcription were analysed. A distinct biphasic cellular response to OP with stimulated gelatinase A protein expression and enzyme activity during the initial 24 h, and subsequent inhibition, was apparent, as shown by gelatin zymography. Gelatinase B activity remained unchanged. The abundance of gelatinase A transcripts, determined by reverse transcriptase-PCR, indicated a concordant regulation of gene transcription. To elucidate underlying regulatory events, reporter constructs were transfected. In these experiments, a recently identified response element, RE-1, conferred a significant stimulatory effect within the initial 4 h of OP. Nuclear protein/RE-1 binding studies revealed additional complexes from 5 min up to 3 h after OP exposure, with intensities dependent on P(max). STAT3 was identified as a component of these novel complexes. Down-regulation of cis-activity after 48 h of OP exposure was not transferred via the proximal 1686 bp of the gelatinase A regulatory sequence. In conclusion, hyperbaric OP elicits time-dependent changes in rat MC gelatinase A gene transcription.


Glomerular Mesangium/enzymology , Matrix Metalloproteinase 2/genetics , Pressure , Transcription, Genetic , Animals , Base Sequence , Cell Nucleus/enzymology , Cells, Cultured , DNA Primers , Gene Expression Regulation, Enzymologic , Hypertension, Renal/enzymology , Kinetics , Matrix Metalloproteinase Inhibitors , Oscillometry , Polymerase Chain Reaction , Rats , Time Factors
...