Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Elife ; 82019 03 22.
Article in English | MEDLINE | ID: mdl-30900988

ABSTRACT

Unraveling the genetic susceptibility of complex diseases such as chronic kidney disease remains challenging. Here, we used inbred rat models of kidney damage associated with elevated blood pressure for the comprehensive analysis of a major albuminuria susceptibility locus detected in these models. We characterized its genomic architecture by congenic substitution mapping, targeted next-generation sequencing, and compartment-specific RNA sequencing analysis in isolated glomeruli. This led to prioritization of transmembrane protein Tmem63c as a novel potential target. Tmem63c is differentially expressed in glomeruli of allele-specific rat models during onset of albuminuria. Patients with focal segmental glomerulosclerosis exhibited specific TMEM63C loss in podocytes. Functional analysis in zebrafish revealed a role for tmem63c in mediating the glomerular filtration barrier function. Our data demonstrate that integrative analysis of the genomic architecture of a complex trait locus is a powerful tool for identification of new targets such as Tmem63c for further translational investigation.


Subject(s)
Genetic Loci , Genetic Predisposition to Disease , Hypertension, Renal/physiopathology , Hypertension/complications , Multifactorial Inheritance , Nephritis/physiopathology , Albuminuria/pathology , Animals , Disease Models, Animal , Humans , Hypertension, Renal/pathology , Nephritis/pathology , Rats , Zebrafish
2.
Ren Fail ; 39(1): 629-642, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28805484

ABSTRACT

Uncontrolled activation of transforming growth factor beta (TGF-ß) family members is hypothesized to participate in type 2 diabetes (T2D) dependent diabetic nephropathy (DN). We evaluated and compared downstream activation of the Smad2-signaling pathway in kidney samples from T2D patients to kidneys from the T2D model of leptin receptor deficient db/db mouse. Furthermore, expression of TGF-ß family members was evaluated to elucidate molecular mechanisms in the mouse model. Kidney samples from patients with advanced stages of DN showed elevated pSmad2 staining whereas db/db mouse kidneys surprisingly showed a decrease in pSmad2 in the tubular compartment. Structurally, kidney tissue showed dilated tubules and expanded glomeruli, but no clear fibrotic pattern was found in the diabetic mice. Selective TGF-ß family members were up-regulated at the mRNA level. Antagonists of bone morphogenetic protein (BMP) ligands, such as Gremlin1, USAG1 and Sclerostin, were strongly up-regulated suggesting a dampening effect on BMP pathways. Together, these results indicate a lack of translation from T2D patient kidneys to the db/db model with regards to Smad signaling pathway. It is plausible that a strong up-regulation of BMP antagonizing factors account for the lack of Smad1/5/8 activation, in spite of increased expression of several BMP members.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/pathology , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Smad2 Protein/metabolism , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/etiology , Disease Models, Animal , Female , Fibrosis , Glycoproteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Phosphorylation , RNA, Messenger/metabolism , Receptors, Leptin/genetics , Signal Transduction , Transforming Growth Factor beta1 , Up-Regulation
3.
Sci Rep ; 7: 44492, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281693

ABSTRACT

We previously demonstrated that polymorphisms in the carnosinase-1 gene (CNDP1) determine the risk of nephropathy in type 2 diabetic patients. Carnosine, the substrate of the enzyme encoded by this gene, is considered renoprotective and could possibly be used to treat diabetic nephropathy (DN). In this study, we examined the effect of carnosine treatment in vivo in BTBR (Black and Tan, BRachyuric) ob/ob mice, a type 2 diabetes model which develops a phenotype that closely resembles advanced human DN. Treatment of BTBR ob/ob mice with 4 mM carnosine for 18 weeks reduced plasma glucose and HbA1c, concomitant with elevated insulin and C-peptide levels. Also, albuminuria and kidney weights were reduced in carnosine-treated mice, which showed less glomerular hypertrophy due to a decrease in the surface area of Bowman's capsule and space. Carnosine treatment restored the glomerular ultrastructure without affecting podocyte number, resulted in a modified molecular composition of the expanded mesangial matrix and led to the formation of carnosine-acrolein adducts. Our results demonstrate that treatment with carnosine improves glucose metabolism, albuminuria and pathology in BTBR ob/ob mice. Hence, carnosine could be a novel therapeutic strategy to treat patients with DN and/or be used to prevent DN in patients with diabetes.


Subject(s)
Albuminuria/diet therapy , Carnosine/pharmacology , Diabetes Mellitus, Type 2/diet therapy , Diabetic Nephropathies/diet therapy , Hypoglycemic Agents/pharmacology , Administration, Oral , Albuminuria/blood , Albuminuria/genetics , Albuminuria/pathology , Animals , Blood Glucose/metabolism , C-Peptide/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/blood , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Dipeptidases/genetics , Dipeptidases/metabolism , Disease Models, Animal , Gene Expression , Glomerular Mesangium/drug effects , Glycated Hemoglobin/genetics , Glycated Hemoglobin/metabolism , Humans , Insulin/blood , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Mice , Mice, Obese , Organ Size/drug effects
4.
Nephron ; 135(4): 291-306, 2017.
Article in English | MEDLINE | ID: mdl-28064277

ABSTRACT

BACKGROUND: The role of transforming growth factor-ß (TGF-ß) has recently gained much attention in diabetic nephropathy and kidney fibrosis. In this study, we extend this to an assessment of transcriptional regulation of the entire TGF-ß superfamily in kidneys from diabetic vs. healthy mice. In order to study the translation between mouse model and patients, we evaluated the signature of phosphorylated Sma- and Mad-related protein 2 (pSmad2), as molecular marker of TGF-ß/activin activity, in the kidneys of streptozotocin (STZ)-treated mice compared to that of type 1 diabetes (T1D) patients. METHODS: Patterns of pSmad2 were determined in kidneys from T1D patients with progressed diabetic nephropathy (DN), defined by hyperglycemia, microalbuminuria, and increased levels of serum creatinine. They were compared to changes seen in the STZ-induced DN mouse model. This was studied by immunohistochemistry (IHC) with an antibody specific for pSmad2. Diabetic mice were also characterized by pSmad1/5/8 (IHC), pSmad2/3 (flow cytometry), and TGF-ß family members including bone morphogenetic protein (BMP)-like proteins (quantitative real-time polymerase chain reaction [qPCR]). RESULTS: Renal tubules in DN patients and in STZ mice showed upregulation of pSmad2 concomitant with significantly enlarged distal tubule lumens (p < 0.0001). Renal-derived CD11b+ cells from STZ mice showed elevated pSmad2/3, while endothelial cells had reduced pSmad2/3 levels. No pSmad1/5/8 was observed in the tubule compartment of STZ-treated mice. On total kidney mRNA level, a signature favoring activation of the TGF-ß/activin pathway and inhibition of the BMP pathway was demonstrated by qPCR. CONCLUSION: Although the pre-clinical DN model lacks the features of fibrosis present in human DN, both species show induction of a local milieu favoring pSmad2 signaling, which may be useful as a disease biomarker in pre-clinical models.


Subject(s)
Diabetic Nephropathies/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Activins/genetics , Adult , Aged , Aged, 80 and over , Animals , Bone Morphogenetic Proteins/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice , Mice, 129 Strain , Middle Aged , Models, Biological , Phosphorylation , Smad2 Protein/blood , Smad3 Protein/blood , Transforming Growth Factor beta/genetics , Up-Regulation
5.
Transplantation ; 101(9): 2017-2025, 2017 09.
Article in English | MEDLINE | ID: mdl-27926596

ABSTRACT

BACKGROUND: Glucocorticoid (GC)-refractory acute rejection (AR) is a risk factor for inferior renal allograft outcome. We investigated genetic predisposition to the response to steroid treatment of acute allograft rejection. METHODS: Single nucleotide polymorphisms of genes involved in GC signaling (GR, GLCCI1) and drug metabolism and transport (CYP3A5, ABCB1, and PXR) were analyzed in kidney transplant recipients (1995-2005, Leiden cohort, n = 153) treated with methylprednisolone. Significant associations were verified in a second cohort (Berlin cohort, n = 66). RESULTS: Patients who received a CYP3A5*1 allele expressing allograft had a lower risk of resistance to methylprednisolone during AR (odds ratio, 0.29; 95% confidence interval, 0.11-0.79; P = 0.016 in combined cohorts analysis). No differences were observed for GC signaling or other drug metabolism/transport-related genes. Both before transplantation (n = 69) and at time of AR (n = 88), tissue CYP3A5 mRNA expression was significantly higher in CYP3A5*1 allele expressing donor kidneys than in CYP3A5*3/*3 allografts (P < 0.00001). Moreover, steroid-responsive patients (n = 64) expressed significantly higher intragraft CYP3A5 mRNA levels compared to steroid-refractory patients (n = 42) in AR (P = 0.006). CONCLUSIONS: CYP3A5 protein expression was detected in tubular epithelial cells and inflammatory cells within the grafts. Our findings show that steroid resistance during AR is associated with donor genotype and intragraft expression levels of CYP3A5.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Glucocorticoids/therapeutic use , Graft Rejection/drug therapy , Kidney Transplantation/adverse effects , Kidney/drug effects , Kidney/surgery , Methylprednisolone/therapeutic use , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Tissue Donors , Acute Disease , Allografts , Chi-Square Distribution , Cytochrome P-450 CYP3A/metabolism , Drug Resistance/genetics , Female , Gene Frequency , Genotype , Germany , Glucocorticoids/metabolism , Graft Rejection/enzymology , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Kaplan-Meier Estimate , Kidney/enzymology , Logistic Models , Male , Methylprednisolone/metabolism , Middle Aged , Netherlands , Odds Ratio , Pharmacogenetics , Pharmacogenomic Testing , Phenotype , Proportional Hazards Models , RNA, Messenger/genetics , Risk Factors , Treatment Outcome
6.
J Pathol ; 241(5): 589-599, 2017 04.
Article in English | MEDLINE | ID: mdl-27976371

ABSTRACT

Diabetic nephropathy is the leading cause of end-stage renal disease. Diabetic patients have increased plasma concentrations of apolipoprotein C-I (apoCI), and meta-analyses found that a polymorphism in APOC1 is associated with an increased risk of developing nephropathy. To investigate whether overexpressing apoCI contributes to the development of kidney damage, we studied renal tissue and peritoneal macrophages from APOC1 transgenic (APOC1-tg) mice and wild-type littermates. In addition, we examined renal material from autopsied diabetic patients with and without diabetic nephropathy and from autopsied control subjects. We found that APOC1-tg mice, but not wild-type mice, develop albuminuria, renal dysfunction, and glomerulosclerosis with increased numbers of glomerular M1 macrophages. Moreover, compared to wild-type macrophages, stimulated macrophages isolated from APOC1-tg mice have increased cytokine expression, including TNF-alpha and TGF-beta, both of which are known to increase the production of extracellular matrix proteins in mesangial cells. These results suggest that APOC1 expression induces glomerulosclerosis, potentially by increasing the cytokine response in macrophages. Furthermore, we detected apoCI in the kidneys of diabetic patients, but not in control kidneys. Moreover, patients with diabetic nephropathy have significantly more apoCI present in glomeruli compared to diabetic patients without nephropathy, suggesting that apoCI could be involved in the development of diabetic nephropathy. ApoCI co-localized with macrophages. Therefore, apoCI is a promising new therapeutic target for patients at risk of developing nephropathy. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Apolipoprotein C-I/metabolism , Diabetic Nephropathies/etiology , Gene Expression Regulation , Kidney Failure, Chronic/etiology , Aged , Albuminuria/etiology , Albuminuria/pathology , Animals , Apolipoprotein C-I/genetics , Brain/metabolism , Brain/pathology , Cytokines/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Female , Humans , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lung/metabolism , Lung/pathology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Pancreas/metabolism , Pancreas/pathology , Spleen/metabolism , Spleen/pathology
7.
Elife ; 52016 Sep 26.
Article in English | MEDLINE | ID: mdl-27668431

ABSTRACT

TFE-fusion renal cell carcinomas (TFE-fusion RCCs) are caused by chromosomal translocations that lead to overexpression of the TFEB and TFE3 genes (Kauffman et al., 2014). The mechanisms leading to kidney tumor development remain uncharacterized and effective therapies are yet to be identified. Hence, the need to model these diseases in an experimental animal system (Kauffman et al., 2014). Here, we show that kidney-specific TFEB overexpression in transgenic mice, resulted in renal clear cells, multi-layered basement membranes, severe cystic pathology, and ultimately papillary carcinomas with hepatic metastases. These features closely recapitulate those observed in both TFEB- and TFE3-mediated human kidney tumors. Analysis of kidney samples revealed transcriptional induction and enhanced signaling of the WNT ß-catenin pathway. WNT signaling inhibitors normalized the proliferation rate of primary kidney cells and significantly rescued the disease phenotype in vivo. These data shed new light on the mechanisms underlying TFE-fusion RCCs and suggest a possible therapeutic strategy based on the inhibition of the WNT pathway.

8.
Am J Nephrol ; 44(3): 194-203, 2016.
Article in English | MEDLINE | ID: mdl-27578560

ABSTRACT

BACKGROUND: In autosomal dominant polycystic kidney disease, renoprotective treatment with a vasopressin V2 receptor antagonist (V2RA) is given in a fixed dose (FD). Disease progression and drug habituation could diminish treatment efficacy. We investigated whether the renoprotective effect of the V2RA can be improved by dose titration of the V2RA aiming to maintain aquaresis at a high level. METHODS: The V2RA OPC-31260 was administered to Pkd1-deletion mice in an FD (0.1%) or in a titrated dose (TD, up to 0.8% when drinking volume dropped). Total kidney weight (TKW) and cyst ratio were investigated and compared to non-treated Pkd1-deletion mice. Treatment was started early or late (21 or 42 days postnatal). RESULTS: Water intake was significantly higher throughout the experiment in the TD compared to the FD group. FD treatment that was initiated early reduced TKW and cyst ratio but lost its renoprotective effect later during the experiment. In contrast, TD treatment was able to maintain the renoprotective effect. TD treatment, however, was also associated with a higher early termination rate in comparison with FD treatment. Late start of treatment (FD or TD) did not show a renoprotective effect. CONCLUSIONS: Titration of a V2RA aimed to maintain aquaresis at a high level showed a better renoprotective effect compared to FD administration. However, this treatment regimen was poorly tolerated and did not overcome treatment unresponsiveness when started later in the disease.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/administration & dosage , Benzazepines/administration & dosage , Kidney/pathology , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/pathology , Animals , Cysts/pathology , Disease Models, Animal , Drinking/drug effects , Female , Male , Mice, Knockout , Organ Size/drug effects , Polycystic Kidney, Autosomal Dominant/genetics , Protein Kinase C/genetics , Time Factors , Water
9.
J Am Soc Nephrol ; 27(12): 3589-3599, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27020852

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD), characterized by the formation of numerous kidney cysts, is caused by PKD1 or PKD2 mutations and affects 0.1% of the population. Although recent clinical studies indicate that reduction of cAMP levels slows progression of PKD, this finding has not led to an established safe and effective therapy for patients, indicating the need to find new therapeutic targets. The role of TGF-ß in PKD is not clearly understood, but nuclear accumulation of phosphorylated SMAD2/3 in cyst-lining cells suggests the involvement of TGF-ß signaling in this disease. In this study, we ablated the TGF-ß type 1 receptor (also termed activin receptor-like kinase 5) in renal epithelial cells of PKD mice, which had little to no effect on the expression of SMAD2/3 target genes or the progression of PKD. Therefore, we investigated whether alternative TGF-ß superfamily ligands account for SMAD2/3 activation in cystic epithelial cells. Activins are members of the TGF-ß superfamily and drive SMAD2/3 phosphorylation via activin receptors, but activins have not been studied in the context of PKD. Mice with PKD had increased expression of activin ligands, even at early stages of disease. In addition, treatment with a soluble activin receptor IIB fusion (sActRIIB-Fc) protein, which acts as a soluble trap to sequester activin ligands, effectively inhibited cyst formation in three distinct mouse models of PKD. These data point to activin signaling as a key pathway in PKD and a promising target for therapy.


Subject(s)
Activins/antagonists & inhibitors , Polycystic Kidney Diseases/prevention & control , Signal Transduction , Animals , Disease Progression , Epithelial Cells , Female , Kidney/cytology , Male , Mice , Polycystic Kidney Diseases/etiology , Recombinant Fusion Proteins/pharmacology , Smad2 Protein/physiology , Smad3 Protein/physiology , Time Factors
10.
Am J Physiol Endocrinol Metab ; 309(7): E663-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26389600

ABSTRACT

A particular allele of the carnosinase gene (CNDP1) is associated with reduced plasma carnosinase activity and reduced risk for nephropathy in diabetic patients. On the one hand, animal and human data suggest that hyperglycemia increases plasma carnosinase activity. On the other hand, we recently reported lower carnosinase activity levels in elite athletes involved in high-intensity exercise compared with untrained controls. Therefore, this study investigates whether exercise training and the consequent reduction in hyperglycemia can suppress carnosinase activity and content in adults with type 2 diabetes. Plasma samples were taken from 243 males and females with type 2 diabetes (mean age = 54.3 yr, SD = 7.1) without major microvascular complications before and after a 6-mo exercise training program [4 groups: sedentary control (n = 61), aerobic exercise (n = 59), resistance exercise (n = 63), and combined exercise training (n = 60)]. Plasma carnosinase content and activity, hemoglobin (Hb) A1c, lipid profile, and blood pressure were measured. A 6-mo exercise training intervention, irrespective of training modality, did not decrease plasma carnosinase content or activity in type 2 diabetic patients. Plasma carnosinase content and activity showed a high interindividual but very low intraindividual variability over the 6-mo period. Age and sex, but not Hb A1c, were significantly related to the activity or content of this enzyme. It can be concluded that the beneficial effects of exercise training on the incidence of diabetic complications are probably not related to a lowering effect on plasma carnosinase content or activity.


Subject(s)
Diabetes Mellitus, Type 2/blood , Dipeptidases/blood , Exercise/physiology , Resistance Training , Adult , Age Factors , Blood Glucose/metabolism , Blood Pressure , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Lipids/blood , Male , Middle Aged , Sex Factors
11.
Amino Acids ; 47(12): 2541-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26206726

ABSTRACT

Histidine-containing dipeptides like carnosine and anserine have protective functions in both health and disease. Animal studies suggest that carnosine can be metabolized within the kidney. The goal of this study was to obtain evidence of carnosine metabolism in the human kidney and to provide insight with regards to diabetic nephropathy. Expression, distribution, and localization of carnosinase-1 (CNDP1), carnosine synthase (CARNS), and taurine transporters (TauT) were measured in human kidneys. CNDP1 and CARNS activities were measured in vitro. CNDP1 and CARNS were located primarily in distal and proximal tubules, respectively. Specifically, CNDP1 levels were high in tubular cells and podocytes (20.3 ± 3.4 and 15 ± 3.2 ng/mg, respectively) and considerably lower in endothelial cells (0.5 ± 0.1 ng/mg). CNDP1 expression was correlated with the degradation of carnosine and anserine (r = 0.88 and 0.81, respectively). Anserine and carnosine were also detectable by HPLC in the renal cortex. Finally, TauT mRNA and protein were found in all renal epithelial cells. In diabetic patients, CNDP1 seemed to be reallocated to proximal tubules. We report compelling evidence that the kidney has an intrinsic capacity to metabolize carnosine. Both CNDP1 and CARNS are expressed in glomeruli and tubular cells. Carnosine-synthesizing and carnosine-hydrolyzing enzymes are localized in distinct compartments in the nephron and increased CNDP1 levels suggest a higher CNDP1 activity in diabetic kidneys.


Subject(s)
Carnosine/metabolism , Gene Expression Regulation , Kidney/metabolism , Anserine/metabolism , Chromatography, High Pressure Liquid , Diabetic Neuropathies/metabolism , Dipeptidases/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Gene Expression Profiling , Humans , Hydrolysis , Immunohistochemistry , Kidney Tubules/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Nephrons/metabolism , Peptide Synthases/metabolism , Podocytes/metabolism , RNA, Messenger/metabolism
12.
J Biol Chem ; 290(28): 17190-205, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26001783

ABSTRACT

Anserine (ß-alanyl-N(Pi)-methyl-L-histidine), a methylated derivative of carnosine (ß-alanyl-L-histidine), is an abundant constituent of vertebrate skeletal muscles. Although it has been suggested to serve as a proton buffer and radical scavenger, its physiological function remains mysterious. The formation of anserine is catalyzed by carnosine N-methyltransferase, recently identified in chicken as histamine N-methyltransferase-like (HNMT-like) protein. Although the HNMT-like gene is absent in mammalian genomes, the activity of carnosine N-methyltransferase was reported in most mammalian species. In the present investigation, we purified carnosine N-methyltransferase from rat muscles about 2600-fold. Three polypeptides of ∼ 45, 50, and 70 kDa coeluting with the enzyme activity were identified in the preparation. Mass spectrometry analysis of these polypeptides resulted in the identification of UPF0586 protein C9orf41 homolog as the only meaningful candidate. Rat UPF0586 and its yeast, chicken, and human orthologs were expressed in COS-7 cells and purified to homogeneity. Although all recombinant proteins catalyzed the formation of anserine, as confirmed by chromatographic and mass spectrometry analysis, rat UPF0586 was more active on carnosine than other orthologs. Confocal microscopy of HeLa cells expressing recombinant UPF5086 proteins revealed their presence in both cytosol and nucleus. Carnosine and Gly-His were the best substrates for all UPF0586 orthologs studied, although the enzymes also methylated other l-histidine-containing di- and tripeptides. Finally, cotransfection of COS-7 cells with rat or human UPF0586 and carnosine synthase transformed the cells into efficient anserine producers. We conclude that UPF0586 is mammalian carnosine N-methyltransferase and hypothesize that it may also serve as a peptide or protein methyltransferase in eukaryotes.


Subject(s)
Anserine/biosynthesis , Protein Methyltransferases/metabolism , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Carnosine/metabolism , Chickens , Chlorocebus aethiops , DNA/genetics , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Muscle, Skeletal/enzymology , Phylogeny , Protein Methyltransferases/chemistry , Protein Methyltransferases/genetics , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Tandem Mass Spectrometry
13.
J Am Soc Nephrol ; 26(6): 1322-33, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25361818

ABSTRACT

In total, 1 in 1000 individuals carries a germline mutation in the PKD1 or PKD2 gene, which leads to autosomal dominant polycystic kidney disease (ADPKD). Cysts can form early in life and progressively increase in number and size during adulthood. Extensive research has led to the presumption that somatic inactivation of the remaining allele initiates the formation of cysts, and the progression is further accelerated by renal injury. However, this hypothesis is primarily on the basis of animal studies, in which the gene is inactivated simultaneously in large percentages of kidney cells. To mimic human ADPKD in mice more precisely, we reduced the percentage of Pkd1-deficient kidney cells to 8%. Notably, no pathologic changes occurred for 6 months after Pkd1 deletion, and additional renal injury increased the likelihood of cyst formation but never triggered rapid PKD. In mildly affected mice, cysts were not randomly distributed throughout the kidney but formed in clusters, which could be explained by increased PKD-related signaling in not only cystic epithelial cells but also, healthy-appearing tubules near cysts. In the majority of mice, these changes preceded a rapid and massive onset of severe PKD that was remarkably similar to human ADPKD. Our data suggest that initial cysts are the principal trigger for a snowball effect driving the formation of new cysts, leading to the progression of severe PKD. In addition, this approach is a suitable model for mimicking human ADPKD and can be used for preclinical testing.


Subject(s)
Gene Deletion , Germ-Line Mutation , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Tamoxifen/adverse effects , Animals , Cell Proliferation , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Phenotype , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Polycystic Kidney, Autosomal Dominant/pathology , Polycystic Kidney, Autosomal Dominant/physiopathology , Random Allocation , Recombination, Genetic , Signal Transduction , Statistics, Nonparametric , Tamoxifen/pharmacology
14.
J Appl Physiol (1985) ; 116(5): 553-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24408989

ABSTRACT

Given the ergogenic properties of ß-alanyl-L-histidine (carnosine) in skeletal muscle, it can be hypothesized that elevated levels of circulating carnosine could equally be advantageous for high-intensity exercises. Serum carnosinase (CN1), the enzyme hydrolyzing the dipeptide, is highly active in the human circulation. Consequently, dietary intake of carnosine usually results in rapid degradation upon absorption, yet this is less pronounced in subjects with low CN1 activity. Therefore, acute carnosine supplementation before high-intensity exercise could be ergogenic in these subjects. In a cross-sectional study, we determined plasma CN1 activity and content in 235 subjects, including 154 untrained controls and 45 explosive and 36 middle- to long-distance elite athletes. In a subsequent double-blind, placebo-controlled, crossover study, 12 men performed a cycling capacity test at 110% maximal power output (CCT 110%) following acute carnosine (20 mg/kg body wt) or placebo supplementation. Blood samples were collected to measure CN1 content, carnosine, and acid-base balance. Both male and female explosive athletes had significantly lower CN1 activity (14% and 21% lower, respectively) and content (30% and 33% lower, respectively) than controls. Acute carnosine supplementation resulted only in three subjects in carnosinemia. The CCT 110% performance was not improved after carnosine supplementation, even when accounting for low/high CN1 content. No differences were found in acid-base balance, except for elevated resting bicarbonate following carnosine supplementation and in low CN1 subjects. In conclusion, explosive athletes have lower serum CN1 activity and content compared with untrained controls, possibly resulting from genetic selection. Acute carnosine supplementation does not improve high-intensity performance.


Subject(s)
Dipeptidases/blood , Exercise/physiology , Acid-Base Equilibrium/physiology , Adolescent , Adult , Bicarbonates/blood , Carnosine/pharmacology , Colorimetry , Creatine Kinase/metabolism , Cross-Over Studies , Cross-Sectional Studies , Dietary Supplements , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Ferric Compounds/blood , Ferric Compounds/metabolism , Humans , Male , Middle Aged , Oxidation-Reduction , Sports , Thiobarbituric Acid Reactive Substances , Young Adult
15.
Kidney Int ; 84(2): 238-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23903419

ABSTRACT

Chaudhary and colleagues describe an in vivo imaging technique for detecting altered glomerular extracellular matrix after development of glomerulonephritis. Using fluorochrome-labeled Fab fragments of the human monoclonal antibody F1.1 against the NC1 domain of the α3 chain of collagen type IV, in vivo binding is shown in glomeruli from mice with nephritis, while the activation of complement and Fc receptors is prevented. This novel method might allow rapid in vivo detection of renal disease.


Subject(s)
Antibodies, Monoclonal , Autoantigens/analysis , Collagen Type IV/analysis , Kidney Glomerulus/chemistry , Nephritis/diagnosis , Nephrosis/diagnosis , Animals , Female , Humans , Male
16.
Transplantation ; 96(3): 282-9, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23839000

ABSTRACT

BACKGROUND: Recent insights suggest that endothelial cell (EC) activation plays a major role in renal ischemia-reperfusion (I/R) injury. Interactions between ECs and pericytes via signaling molecules, including angiopoietins, are involved in maintenance of the vascular integrity. Experimental data have shown that enhancement of Angiopoietin (Ang)-1 signaling might be beneficial in renal I/R injury. However, little is known about the role of angiopoietins in human renal I/R injury. METHODS: In this study, EC activation and changes in angiopoeitins are assessed in human living-donor (LD) and deceased-donor (DD) kidney transplantation. Local release of angiopoietins was measured by unique, dynamic arteriovenous measurements over the reperfused kidney. RESULTS: Renal I/R is associated with acute EC activation shown by a vast Ang-2 release from both LD and DD shortly after reperfusion. Its counterpart Ang-1 was not released. Histologic analysis of kidney biopsies showed EC loss after reperfusion. Baseline protein and mRNA Ang-1 expression was significantly reduced in DD compared with LD and declined further after reperfusion. CONCLUSIONS: Human renal I/R injury induces EC activation after reperfusion reflected by Ang-2 release from the kidney. Interventions aimed at maintenance of vascular integrity by modulating angiopoietin signaling may be promising in human clinical kidney transplantation.


Subject(s)
Angiopoietin-2/physiology , Kidney Transplantation , Kidney/blood supply , Living Donors , Reperfusion Injury/physiopathology , Angiopoietin-1/physiology , Humans , von Willebrand Factor/physiology
17.
Kidney Int ; 83(6): 1099-108, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23466997

ABSTRACT

Autosomal-dominant polycystic kidney disease is characterized by progressive cyst formation and fibrosis in the kidneys. Here we describe an orthologous Pkd1(nl,nl) mouse model, with reduced expression of the normal Pkd1 transcript, on a fixed genetic background of equal parts C57Bl/6 and 129Ola/Hsd mice (B6Ola-Pkd1(nl,nl)). In these mice, the first cysts develop from mature proximal tubules around birth. Subsequently, larger cysts become visible at day 7, followed by distal tubule and collecting duct cyst formation, and progressive cystic enlargement to develop into large cystic kidneys within 4 weeks. Interestingly, cyst expansion was followed by renal volume regression due to cyst collapse. This was accompanied by focal formation of fibrotic areas, an increased expression of genes involved in matrix remodeling and subsequently an increase in infiltrating immune cells. After an initial increase in blood urea within the first 4 weeks, renal function remained stable over time and the mice were able to survive up to a year. Also, in kidneys of ADPKD patients collapsed cysts were observed, in addition to massive fibrosis and immune infiltrates. Thus, B6Ola-Pkd1(nl,nl) mice show regression of cysts and renal volume that is not accompanied by a reduction in blood urea levels.


Subject(s)
Kidney/pathology , Polycystic Kidney, Autosomal Dominant/pathology , Age Factors , Animals , Biomarkers/blood , Disease Models, Animal , Disease Progression , Extracellular Matrix/metabolism , Fibrosis , Gene Expression Regulation , Humans , Kidney/immunology , Kidney/metabolism , Kidney/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Polycystic Kidney, Autosomal Dominant/blood , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/immunology , Polycystic Kidney, Autosomal Dominant/physiopathology , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Urea/blood
18.
Transplantation ; 94(9): 903-10, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23047455

ABSTRACT

BACKGROUND: Allogeneic islets of Langerhans transplantation is hampered in its success as a curative treatment of type 1 diabetes by the absence of potent, specific, and nontoxic immunosuppressive drugs. Here, we assessed whether donor bone marrow-derived dexamethasone-treated dendritic cells (dexDCs) could prolong islet allograft survival in a full major histocompatibility complex mismatch rat model. METHODS: Rodent allogeneic islet transplantation was performed from DA rats to Lewis rats and vice versa. Permanently immature dendritic cells were generated from the bone marrow of DA and Lewis rats by treatment with dexamethasone. Animals were either vehicle or donor dexDCs pretreated. Serum was used to monitor glucose, C-peptide, and alloreactive antibodies. RESULTS: The transplantation of DA islets into Lewis recipients showed direct graft failure with reduced numbers of ß-cells when rats were pretreated with donor dexDCs. In the reverse model (Lewis islets into DA recipients), dexDC-treated DA recipients even showed a significantly accelerated rejection of Lewis islets. Immunohistochemical analysis of allograft tissue of dexDC-treated recipients showed a predominant natural killer cell infiltration and a presence of antibody reactivity in the absence of complement deposition. Alloreactive antibodies were solely found in dexDC-treated recipients. CONCLUSION: Our study shows that pretreatment with donor-derived dexDCs induces an antibody-mediated rejection in this islet transplantation rodent model.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/transplantation , Dexamethasone/pharmacology , Diabetes Mellitus, Experimental/surgery , Graft Rejection/immunology , Islets of Langerhans Transplantation/immunology , Animals , Antibodies/blood , Cell Movement/physiology , Dendritic Cells/pathology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Female , Killer Cells, Natural/pathology , Male , Rats , Rats, Inbred Lew , Rats, Inbred Strains , Streptozocin/adverse effects , T-Lymphocytes/pathology , Time Factors
19.
Am J Physiol Renal Physiol ; 302(12): F1537-44, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22496410

ABSTRACT

A polymorphism in the carnosine dipeptidase-1 gene (CNDP1), resulting in decreased plasma carnosinase activity, is associated with a reduced risk for diabetic nephropathy. Because carnosine, a natural scavenger/suppressor of ROS, advanced glycation end products, and reactive aldehydes, is readily degraded in blood by the highly active carnosinase enzyme, it has been postulated that low serum carnosinase activity might be advantageous to reduce diabetic complications. The aim of this study was to examine whether low carnosinase activity promotes circulating carnosine levels after carnosine supplementation in humans. Blood and urine were sampled in 25 healthy subjects after acute supplementation with 60 mg/kg body wt carnosine. Precooled EDTA-containing tubes were used for blood withdrawal, and plasma samples were immediately deproteinized and analyzed for carnosine and ß-alanine by HPLC. CNDP1 genotype, baseline plasma carnosinase activity, and protein content were assessed. Upon carnosine ingestion, 8 of the 25 subjects (responders) displayed a measurable increase in plasma carnosine up to 1 h after supplementation. Subjects with no measurable increment in plasma carnosine (nonresponders) had ∼2-fold higher plasma carnosinase protein content and ∼1.5-fold higher activity compared with responders. Urinary carnosine recovery was 2.6-fold higher in responders versus nonresponders and was negatively dependent on both the activity and protein content of the plasma carnosinase enzyme. In conclusion, low plasma carnosinase activity promotes the presence of circulating carnosine upon an oral challenge. These data may further clarify the link among CNDP1 genotype, carnosinase, and diabetic nephropathy.


Subject(s)
Carnosine/administration & dosage , Dipeptidases/blood , Administration, Oral , Adult , Chromatography, High Pressure Liquid , Diabetic Nephropathies/genetics , Dipeptidases/genetics , Dipeptidases/urine , Female , Humans , Male , beta-Alanine/blood
20.
PLoS One ; 7(3): e32347, 2012.
Article in English | MEDLINE | ID: mdl-22479322

ABSTRACT

In pregnancies achieved after egg donation (ED) tolerance towards a completely allogeneic fetus is mediated by several complex immunoregulatory mechanisms, of which numerous aspects are still unknown. A distinct lesion not described previously in the literature, was repeatedly found in the chorionic plate in a substantial portion of placentas from ED pregnancies, but never in placentas from normal term pregnancies. The aim of this study was to assess its origin and its cellular composition. The relation between the lesion, the clinical and histological parameters were assessed. In addition we investigated the relation with the number of HLA-mismatches and KIR genotype of mother and child.In ten out of twenty-six (38.5%) placentas from ED pregnancies an inflammatory lesion was present in the chorionic plate. A significantly lower incidence of pre-eclampsia was found in the group with the lesion; 0% versus 45.5%. A significant relation was found between this lesion and the presence of intervillositis, chronic deciduitis, presence of plasma cells and fibrin deposition in the decidua. Fluorescent in situ hybridisation with X/Y-chromosome probes showed that the majority of cells present in the lesion are of maternal origin. The expression of the macrophage marker CD14+ and of the type 2 macrophage (M2) marker CD163+ was significantly higher in the lesion. The incidence of a fetal HLA-C2 genotype was significantly higher in cases with a lesion compared to the group without the lesion. In conclusion, a striking relationship was observed between the presence of a not previously described inflammatory lesion in the chorionic plate and the absence of pre-eclampsia in ED pregnancies. The lesion consists of mainly maternal cells with a higher expression of the macrophage marker CD14+ and the M2 marker CD163+. These findings suggest a protective immune mechanism which might contribute to the prevention of severe clinical complications like pre-eclampsia.


Subject(s)
Chorion/immunology , Oocyte Donation , Placenta/immunology , Pregnancy Complications/immunology , Adult , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/immunology , Antigens, Differentiation, Myelomonocytic/metabolism , Chorion/metabolism , Chorion/pathology , Decidua/immunology , Decidua/metabolism , Decidua/pathology , Female , Fetus/immunology , Fetus/metabolism , Genotype , HLA-C Antigens/genetics , HLA-C Antigens/immunology , Humans , Immune Tolerance/immunology , Immunohistochemistry , In Situ Hybridization, Fluorescence , Infant, Newborn , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharide Receptors/immunology , Lipopolysaccharide Receptors/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Middle Aged , Placenta/metabolism , Placenta/pathology , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Complications/pathology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Receptors, KIR/genetics , Receptors, KIR/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...