Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Sci Rep ; 13(1): 19660, 2023 11 11.
Article in English | MEDLINE | ID: mdl-37952029

ABSTRACT

Functional and structural alterations of peritubular capillaries (PTCs) are a major determinant of chronic kidney disease (CKD). Using a software-based algorithm for semiautomatic segmentation and morphometric quantification, this study analyzes alterations of PTC shape associated with chronic tubulointerstitial injury in three mouse models and in human biopsies. In normal kidney tissue PTC shape was predominantly elongated, whereas the majority of PTCs associated with chronic tubulointerstitial injury had a rounder shape. This was reflected by significantly reduced PTC luminal area, perimeter and diameters as well as by significantly increased circularity and roundness. These morphological alterations were consistent in all mouse models and human kidney biopsies. The mean circularity of PTCs correlated significantly with categorized glomerular filtration rates and the degree of interstitial fibrosis and tubular atrophy (IFTA) and classified the presence of CKD or IFTA. 3D reconstruction of renal capillaries revealed not only a significant reduction, but more importantly a substantial simplification and reconfiguration of the renal microvasculature in mice with chronic tubulointerstitial injury. Computational modelling predicted that round PTCs can deliver oxygen more homogeneously to the surrounding tissue. Our findings indicate that alterations of PTC shape represent a common and uniform reaction to chronic tubulointerstitial injury independent of the underlying kidney disease.


Subject(s)
Kidney Transplantation , Renal Insufficiency, Chronic , Humans , Mice , Animals , Kidney Tubules/pathology , Capillaries/pathology , Kidney/pathology , Renal Insufficiency, Chronic/pathology , Fibrosis
2.
Kidney Int ; 102(4): 686-688, 2022 10.
Article in English | MEDLINE | ID: mdl-36150758

ABSTRACT

Labes et al. analyze the phosphoproteome in a mouse model of chronic cyclosporine A nephrotoxicity and detect significant changes in the angiogenic pathway. Furthermore, they observe reduced hemoglobin levels and capillary rarefaction in the kidney. The authors show that coadministration of the hypoxia-inducible factor prolyl hydroxylase inhibitor daprodustat almost completely prevents changes of the phosphoproteome and capillary rarefaction, suggesting that prolyl hydroxylase domain enzyme inhibitors may preserve microvasculature of the kidney, which is commonly impaired in chronic kidney disease.


Subject(s)
Microvascular Rarefaction , Prolyl-Hydroxylase Inhibitors , Renal Insufficiency, Chronic , Animals , Cyclosporine , Hemoglobins , Hypoxia-Inducible Factor-Proline Dioxygenases , Mice , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Prolyl-Hydroxylase Inhibitors/therapeutic use , Renal Insufficiency, Chronic/drug therapy
4.
Cell Tissue Res ; 381(1): 125-140, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32189058

ABSTRACT

The role of the hypoxia-inducible transcription factor (HIF) pathway in renal lipid metabolism is largely unknown. As HIF stabilizing prolyl hydroxylase (PHD) inhibitors are currently investigated in clinical trials for the treatment of renal anemia, we studied the effects of genetic deletion and pharmacological inhibition of PHDs on renal lipid metabolism in transgenic mice and human primary tubular epithelial cells (hPTEC). Tubular cell-specific deletion of HIF prolyl hydroxylase 2 (Phd2) increased the size of Oil Red-stained lipid droplets in mice. In hPTEC, the PHD inhibitors (PHDi) DMOG and ICA augmented lipid accumulation, which was visualized by Oil Red staining and assessed by microscopy and an infrared imaging system. PHDi-induced lipid accumulation required the exogenous availability of fatty acids and was observed in both proximal and distal hPTEC. PHDi treatment was not associated with structural features of cytotoxicity in contrast to treatment with the immunosuppressant cyclosporine A (CsA). PHDi and CsA differentially upregulated the expression of the lipid droplet-associated genes PLIN2, PLIN4 and HILPDA. Both PHDi and CsA activated AMP-activated protein kinase (AMPK) indicating the initiation of a metabolic stress response. However, only CsA triggered endoplasmic reticulum (ER) stress as determined by the increased mRNA expression of multiple ER stress markers but CsA-induced ER stress was not linked to lipid accumulation. Our data raise the possibility that PHD inhibition may protect tubular cells from toxic free fatty acids by trapping them as triacylglycerides in lipid droplets. This mechanism might contribute to the renoprotective effects of PHDi in experimental kidney diseases.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Kidney Diseases/drug therapy , Lipid Metabolism/drug effects , Prolyl-Hydroxylase Inhibitors , Animals , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/drug effects , Humans , Kidney Tubules/cytology , Kidney Tubules/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prolyl-Hydroxylase Inhibitors/pharmacology , Prolyl-Hydroxylase Inhibitors/therapeutic use
5.
Sci Rep ; 9(1): 11875, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31417141

ABSTRACT

Non-professional phagocytosis by cancer cells has been described for decades. Recently, non-professional phagocytosis by normal tissue cells has been reported, which prompted us to take a closer look at this phenomenon. Non-professional phagocytosis was studied by staining cultured cells with live-cell staining dyes or by staining paraffin-embedded tissues by immunohistochemistry. Here, we report that each of 21 normal tissue cell lines from seven different organs was capable of phagocytosis, including ex vivo cell cultures examined before the 3rd passage as well as the primary and virus-transformed cell lines. We extended our analysis to an in vivo setting, and we found the occurrence of non-professional phagocytosis in healthy skin biopsies immediately after resection. Using dystrophin immunohistochemistry for membrane staining, human post-infarction myocardial tissue was assessed. We found prominent signs of non-professional phagocytosis at the transition zone of healthy and infarcted myocardia. Taken together, our findings suggest that non-professional phagocytosis is a general feature of normal tissue cells.

6.
Kidney Int ; 96(2): 378-396, 2019 08.
Article in English | MEDLINE | ID: mdl-31146971

ABSTRACT

Prolyl hydroxylase domain enzyme inhibitors (PHDIs) stabilize hypoxia-inducible factors (HIFs), and are protective in models of acute ischemic and inflammatory kidney disease. Whether PHDIs also confer protection in chronic inflammatory kidney disease models remains unknown. Here we investigated long-term effects of PHDI treatment in adenine-induced nephropathy as a model for chronic tubulointerstitial nephritis. After three weeks, renal dysfunction and tubulointerstitial damage, including proximal and distal tubular injury, tubular dilation and renal crystal deposition were significantly attenuated in PHDI-treated (the isoquinoline derivative ICA and Roxadustat) compared to vehicle-treated mice with adenine-induced nephropathy. Crystal-induced renal fibrosis was only partially diminished by treatment with ICA. Renoprotective effects of ICA treatment could not be attributed to changes in adenine metabolism or urinary excretion of the metabolite 2,8-dihydroxyadenine. ICA treatment reduced inflammatory infiltrates of F4/80+ mononuclear phagocytes in the kidneys and supported a regulatory, anti-inflammatory immune response. Furthermore, interstitial deposition of complement C1q was decreased in ICA-treated mice fed an adenine-enriched diet. Tubular cell-specific HIF-1α and myeloid cell-specific HIF-1α and HIF-2α expression were not required for the renoprotective effects of ICA. In contrast, depletion of mononuclear phagocytes with clodronate largely abolished the nephroprotective effects of PHD inhibition. Thus, our findings indicate novel and potent systemic anti-inflammatory properties of PHDIs that confer preservation of kidney function and structure in chronic tubulointerstitial inflammation and might counteract kidney disease progression.


Subject(s)
Nephritis, Interstitial/drug therapy , Phagocytes/drug effects , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Renal Insufficiency, Chronic/prevention & control , Adenine/metabolism , Adenine/toxicity , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Clodronic Acid/pharmacology , Complement C1q/immunology , Complement C1q/metabolism , Disease Models, Animal , Glycine/analogs & derivatives , Glycine/pharmacology , Glycine/therapeutic use , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/immunology , Kidney Tubules/pathology , Male , Mice , Mice, Transgenic , Nephritis, Interstitial/blood , Nephritis, Interstitial/chemically induced , Nephritis, Interstitial/immunology , Phagocytes/immunology , Prolyl Hydroxylases/immunology , Prolyl-Hydroxylase Inhibitors/therapeutic use , Protective Agents/pharmacology , Protective Agents/therapeutic use , Renal Insufficiency, Chronic/immunology
7.
Cell Tissue Res ; 374(3): 619-627, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30078103

ABSTRACT

Renal tubular epithelial cells actively contribute to the development of renal fibrosis and may be targeted by anti-fibrotic drugs. Relaxin-2 (RLX2) applied as recombinant protein is suggested to be renoprotective. Therefore, we investigated whether human primary tubular epithelial cells (hPTEC) obtained from various donors were target cells for the anti-fibrotic actions of RLX2. Treatment of hPTEC with RLX2 reduced the TGF-ß1-induced secretion of the pro-fibrotic factor CTGF (connective tissue growth factor) and inhibited fibronectin synthesis and secretion. Furthermore, metalloproteinase MMP2 secretion was increased, with no effect on MMP9. Considerable differences were observed between hPTEC obtained from different donors. Therefore, expression of the relaxin family peptide receptor RXFP1, the major mediator of renal RLX2 effects, was analyzed. A validated antibody detected a double band of 80-90 kDa in cellular homogenates by Western blotting. Expression of the detected protein was not altered by incubation with TGF-ß1 and RLX2-induced modulation of CTGF expression did not correlate with the putative receptor expression. Therefore, relaxin family receptors RXFP1-4 were assessed by RNA-seq analysis. No evidence was found for mRNA expression of any of these receptors in several hPTEC preparations. Lack of RXFP1 mRNA was confirmed by qPCR using mRNA obtained from THP-1 cells as positive control. Our data thus provide evidence for primary renal human tubular epithelial cells as targets for the anti-fibrotic actions of RLX2. However, anti-fibrotic effects were observed at micromolar concentrations of RLX2 and shown to be independent of RXFP1 expression.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , Kidney Tubules, Proximal/pathology , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Relaxin/pharmacology , Signal Transduction , Transforming Growth Factor beta/metabolism , Cells, Cultured , Connective Tissue Growth Factor/metabolism , Epithelial Cells/drug effects , Fibronectins/metabolism , Fibrosis , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics
8.
Sci Rep ; 8(1): 9497, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29934555

ABSTRACT

Acute kidney injury (AKI) is a common and potentially lethal complication in the hospitalized patients, with hypoxic injury being as a major cause. The loss of renal tubular epithelial cells (TEC), one of the AKI hallmarks, is potentially followed by tubular regeneration process orchestrated by the remaining uninjured TECs that undergo proliferation and migration. In this study, we used human primary TEC to investigate the initiation of tubular cell migration and associated cytoskeletal alterations in response to pharmacological HIF stabilization which resembles the pathophysiology of hypoxia. Tubular cells have been shown to migrate as cohorts in a wound healing assay. Importantly, cells of distal tubular origin moved faster than those of proximal origin. HIF stabilization impaired TEC migration, which was confirmed by live single cell tracking. HIF stabilization significantly reduced tubular cell migration velocity and promoted cell spreading. In contrast to the control conditions, HIF stabilization induced actin filaments rearrangement and cell adhesion molecules including paxillin and focal adhesion kinase. Condensed bundling of keratin fibers was also observed, while the expression of different types of keratins, phosphorylation of keratin 18, and the microtubule structure were not altered. In summary, HIF stabilization reduced the ability of renal tubular cells to migrate and led to cytoskeleton reorganization. Our data suggested an important involvement of HIF stabilization during the epithelial migration underlying the mechanism of renal regeneration in response to AKI.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement , Cytoskeleton/metabolism , Epithelial Cells/cytology , Amino Acids, Dicarboxylic/pharmacology , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Cytoskeleton/drug effects , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Keratins/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Phosphorylation/drug effects , Protein Stability
9.
J Histochem Cytochem ; 65(12): 743-755, 2017 12.
Article in English | MEDLINE | ID: mdl-29058957

ABSTRACT

Connective tissue growth factor (CTGF, also named CCN2) plays an important role in the development of tubulointerstitial fibrosis, which most critically determines the progression to end-stage renal failure in autosomal-dominant polycystic kidney disease (ADPKD), the most common genetically caused renal disease. We determined CTGF expression in a well-characterized animal model of human ADPKD, the PKD/Mhm (cy/+) rat. Kidneys of 12 weeks old (cy/+) as well as (+/+) non-affected rats were analyzed for CTGF RNA and protein expression by RT-PCR, Northern and Western blot analyses, in situ hybridization, and IHC. Besides the established expression of CTGF in glomerular cells in kidneys of wild-type (+/+) animals, in (cy/+) rats, CTGF mRNA and protein were robustly expressed in interstitial, stellate-shaped cells, located in a scattered pattern underlying the cystic epithelium and in focal areas of advanced tubulointerstitial remodeling. Renal CTGF mRNA and protein expression levels were significantly higher in (cy/+) rats compared with their (+/+) littermates. Detection of CTGF expression in cells adjacent to cystic epithelium and in areas of marked fibrosis suggests a role in the local response to cyst development and indicates that CTGF may be a relevant factor contributing to tubulointerstitial fibrosis in polycystic kidney disease.


Subject(s)
Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Gene Expression Regulation , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Animals , Disease Models, Animal , Fibrosis , Kidney/metabolism , Kidney/pathology , Male , Polycystic Kidney, Autosomal Dominant/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
10.
PLoS Genet ; 13(7): e1006872, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28715484

ABSTRACT

Un-physiological activation of hypoxia inducible factor (HIF) is an early event in most renal cell cancers (RCC) following inactivation of the von Hippel-Lindau tumor suppressor. Despite intense study, how this impinges on cancer development is incompletely understood. To test for the impact of genetic signals on this pathway, we aligned human RCC-susceptibility polymorphisms with genome-wide assays of HIF-binding and observed highly significant overlap. Allele-specific assays of HIF binding, chromatin conformation and gene expression together with eQTL analyses in human tumors were applied to mechanistic analysis of one such overlapping site at chromosome 12p12.1. This defined a novel stage-specific mechanism in which the risk polymorphism, rs12814794, directly creates a new HIF-binding site that mediates HIF-1α isoform specific upregulation of its target BHLHE41. The alignment of multiple sites in the HIF cis-acting apparatus with RCC-susceptibility polymorphisms strongly supports a causal model in which minor variation in this pathway exerts significant effects on RCC development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Polymorphism, Single Nucleotide , Alleles , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Renal Cell/diagnosis , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromosomes, Human, Pair 12/genetics , Cyclin D1 , Genome-Wide Association Study , HeLa Cells , Hep G2 Cells , High-Throughput Nucleotide Sequencing , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MCF-7 Cells , Quantitative Trait Loci , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Up-Regulation
11.
Cell Signal ; 29: 31-40, 2017 01.
Article in English | MEDLINE | ID: mdl-27721022

ABSTRACT

Changes in cell morphology that involve alterations of the actin cytoskeleton are a hallmark of diseased renal tubular epithelial cells. While the impact of actin remodeling on gene expression has been analyzed in many model systems based on cell lines, this study investigated human primary tubular epithelial cells isolated from healthy parts of tumor nephrectomies. Latrunculin B (LatB) and cytochalasin D (CytoD) were used to modulate G-actin levels in a receptor-independent manner. Both compounds (at 0.5µM) profoundly altered F-actin structures in a Rho kinase-dependent manner, but only CytoD strongly induced the pro-fibrotic factor CTGF (connective tissue growth factor). CTGF induction was dependent on YAP as shown by transient downregulation experiments. However, CytoD did not alter the nuclear localization of either YAP or TAZ, whereas LatB reduced nuclear levels particularly of TAZ. CytoD modified MKL1, a coactivator of serum response factor (SRF) regulating CTGF induction, and promoted its nuclear localization. TGFß-1 is one of the major factors involved in tubulointerstitial disease and an inducer of CTGF. Preincubation with CytoD but not LatB synergistically enhanced the TGFß-1-stimulated synthesis of CTGF, both in cells cultured on plastic dishes as well as in polarized epithelial cells. CytoD had no direct effect on the phosphorylation of Smad2/3, but facilitated their phosphorylation and thus activation by TGFß-1. Our present findings provide evidence that morphological alterations have a strong impact on cellular signaling of one of the major pro-fibrotic factors, TGFß-1. However, our data also indicate that changes in cell morphology per se cannot predict those interactions which are critically dependent on molecular fine tuning of actin reorganization.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Connective Tissue Growth Factor/metabolism , Cytochalasin D/pharmacology , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Smad Proteins/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta1/pharmacology , Actins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Compartmentation , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Humans , Kidney/cytology , Thiazolidines/pharmacology , Transcription Factors , Transcription, Genetic/drug effects , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , rho-Associated Kinases/metabolism
12.
Purinergic Signal ; 12(4): 687-695, 2016 12.
Article in English | MEDLINE | ID: mdl-27565965

ABSTRACT

Polycystic kidney diseases are characterized by numerous renal cysts that continuously enlarge resulting in compression of intact nephrons and tissue hypoxia. Recently, we have shown that hypoxia-inducible factor (HIF)-1α promotes secretion-dependent cyst expansion, presumably by transcriptional regulation of proteins that are involved in calcium-activated chloride secretion. Here, we report that HIF-1α directly activates expression of the purinergic receptor P2Y2R in human primary renal tubular cells. In addition, we found that P2Y2R is highly expressed in cyst-lining cells of human ADPKD kidneys as well as PKD1 orthologous mouse kidneys. Knockdown of P2Y2R in renal collecting duct cells inhibited calcium-dependent chloride secretion in Ussing chamber analyses. In line with these findings, knockdown of P2Y2R retarded cyst expansion in vitro and prevented ATP- and HIF-1α-dependent cyst growth. In conclusion, P2Y2R mediates ATP-dependent cyst growth and is transcriptionally regulated by HIF-1α. These findings provide further mechanistic evidence on how hypoxia promotes cyst growth.


Subject(s)
Cysts/metabolism , Epithelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Tubules, Proximal/metabolism , Receptors, Purinergic P2Y2/metabolism , Animals , Cysts/pathology , Epithelial Cells/cytology , Female , Humans , Kidney Tubules, Proximal/cytology , Male , Mice , Mice, Knockout , Middle Aged
13.
Biochim Biophys Acta ; 1863(8): 2027-36, 2016 08.
Article in English | MEDLINE | ID: mdl-27155083

ABSTRACT

Pharmacological inhibition of oxygen sensing prolyl hydroxylase domain enzymes (PHDs) has been shown to preserve renal structure and function in various models of kidney disease. Since transforming growth factor ß-1 (TGFß-1) is one of the major mediators of kidney injury, we investigated if inhibition of PHDs with subsequent stabilization of hypoxia inducible transcription factors (HIF) might interfere with TGFß-1 signaling with special emphasis on its target gene connective tissue growth factor (CTGF). Overnight incubation of human renal tubular cells, primary cells and cell lines, with the PDH inhibitor DMOG increased Smad3 expression, but barely affected Smad2. Both Smads were translocated into the nucleus upon activation of the cells with TGFß-1. Interestingly, Smad3 nuclear localization was enhanced upon pretreatment of the cells with DMOG for several hours, whereas nuclear Smad2 was reduced. This differential localization was independent of Smad2/3 phosphorylation. Reduced nuclear Smad2 correlated with impaired CTGF secretion in DMOG-treated cells and transient downregulation of Smad2 interfered with TGFß-1-induced CTGF synthesis. Furthermore, YAP was confirmed as indispensable transcription factor involved in CTGF synthesis. Nuclear localization of YAP and TAZ was reduced in DMOG-treated cells. Our data thus provide evidence for DMOG-mediated reduction of CTGF expression by regulating the nuclear localization of the transcription factors Smad2, YAP and TAZ. Prolonged inhibition of PHDs was necessary to achieve alterations in cellular localization suggesting an indirect HIF-mediated effect. This mechanism might be extended to other transcription factors and target genes, and may thus represent a novel mechanism of negative regulation of gene expression by PHD inhibition.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Connective Tissue Growth Factor/biosynthesis , Kidney Tubules/metabolism , Phosphoproteins/metabolism , Prolyl Hydroxylases/physiology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Amino Acids, Dicarboxylic/pharmacology , Cell Hypoxia/genetics , Cells, Cultured , Connective Tissue Growth Factor/antagonists & inhibitors , Connective Tissue Growth Factor/physiology , Gene Expression Regulation/drug effects , Humans , Kidney Tubules/cytology , Oxygen/metabolism , Phosphoproteins/genetics , Primary Cell Culture , RNA Interference , RNA, Small Interfering/genetics , Smad2 Protein/genetics , Transforming Growth Factor beta1/physiology , YAP-Signaling Proteins
14.
Nanotoxicology ; 10(5): 557-66, 2016.
Article in English | MEDLINE | ID: mdl-26468004

ABSTRACT

Superparamagnetic iron oxide nanoparticles (SPIONs) are in use for many clinical diagnostic and experimental therapeutic applications, for example, for targeted drug delivery. To analyze the cellular responses to mitoxantrone-carrying SPIONs (SPION-MTO), and to the drug released from SPIONs, we used an in vitro system that allows comparison of primary human cells with different endocytotic capacities, namely, epithelial cells from proximal and distal parts of the nephron. SPIONs were selectively and rapidly internalized by proximal tubular cells with high endocytotic potential, but not by distal tubular cells. Uptake did not affect cell viability or morphology. In both cell types, free MTO (10-100 nM) induced double-strand DNA breaks and senescence, cell hypertrophy and reduced cell proliferation. However, cadherin-mediated cell-cell adhesion, cytoskeletal structures or polarity of the cells were not affected. Interestingly, a comparable response was also observed upon treatment with SPION-MTO and was independent of uptake of the particles. The effect of SPION-MTO on cells which did not internalize particles was primarily related to the release of MTO from drug-coated particles upon incubation in serum-containing cell growth medium. In conclusion, we show that whereas the uptake of SPIONs does not affect cellular functions or viability, the toxicity of drug-loaded SPIONs depends essentially on the type of drug bound to nanoparticles. Due to the relatively low systemic toxicity of MTO, the effects of MTO-SPIONs on human tubular cells were moderate, but they may become clinically relevant when more nephrotoxic drugs are bound to SPIONs.


Subject(s)
Antineoplastic Agents/toxicity , Drug Carriers/toxicity , Epithelial Cells/drug effects , Magnetite Nanoparticles/toxicity , Mitoxantrone/toxicity , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cellular Senescence/drug effects , Drug Carriers/chemistry , Drug Liberation , Endocytosis/drug effects , Epithelial Cells/diagnostic imaging , Epithelial Cells/metabolism , Humans , Kidney Tubules/cytology , Magnetite Nanoparticles/chemistry , Mitoxantrone/administration & dosage , Mitoxantrone/chemistry , Primary Cell Culture
15.
PLoS One ; 10(3): e0121589, 2015.
Article in English | MEDLINE | ID: mdl-25816094

ABSTRACT

Morphological alterations of cells can lead to modulation of gene expression. An essential link is the MKL1-dependent activation of serum response factor (SRF), which translates changes in the ratio of G- and F-actin into mRNA transcription. SRF activation is only partially characterized in non-transformed epithelial cells. Therefore, the impact of GTPases of the Rho family and changes in F-actin structures were analyzed in renal proximal tubular epithelial cells. Activation of SRF signaling was compared to the regulation of a known MKL1/SRF target gene, connective tissue growth factor (CTGF). In the human proximal tubular cell line HKC-8 overexpression of two actin mutants either favoring or preventing the formation of F-actin fibers regulated SRF-mediated transcription as well as CTGF expression. Only overexpression of constitutively active RhoA activated SRF-dependent gene expression whereas no effect was detected upon overexpression of Rac1 mutants. To elucidate the functional role of Rho kinases as downstream mediators of RhoA, pharmacological inhibition and genetic inhibition by transient siRNA knock down were compared. Upon stimulation with lysophosphatidic acid (LPA) Rho kinase inhibitors partially suppressed SRF-mediated transcription, whereas interference with Rho kinase expression by siRNA reduced activation of SRF, but barely affected CTGF expression. Together with the partial inhibition of CTGF expression by the pharmacological inhibitors Y27432 and H1154, Rho kinases seem to be less important in mediating RhoA signaling related to CTGF expression in HKC-8 epithelial cells. Short term pharmacological inhibition of Rac1 activity by EHT1864 reduced SRF-dependent CTGF expression in HKC-8 cells, but was overcome by a stimulatory effect after prolonged incubation after 4-6 h. Similarly, human primary cells of proximal but not of distal tubular origin showed inhibitory as well as stimulatory effects of Rac1 inhibition. Thus, RhoA signaling activates MKL1-SRF-mediated CTGF expression in proximal tubular cells, whereas Rac1 signaling is more complex with adaptive cellular responses.


Subject(s)
Actins/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Cells, Cultured , Connective Tissue Growth Factor/metabolism , Epithelial Cells/cytology , Humans , Kidney Tubules, Proximal/cytology , Lysophospholipids/pharmacology , Mutation , RNA, Small Interfering/pharmacology , Serum Response Factor/metabolism , Signal Transduction/drug effects , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics
16.
Int J Biochem Cell Biol ; 46: 39-48, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24275091

ABSTRACT

Src kinases are regulators of the expression of connective tissue growth factor (CTGF/CCN2), which plays a role in fibrotic injuries. The aim of the present study was to evaluate the potential of SU6656, a dual inhibitor of Src family and Aurora kinases, to interfere with the synthesis of this pro-fibrotic factor. SU6656 impaired TGF-ß-mediated upregulation of CTGF mRNA and protein in proximal epithelial HKC-8 cells, and also reduced CTGF expression in cells exposed to autocrine growth factors. In association with the inhibition of Src family kinases and diminished focal adhesion kinase activity, adherence of the cells was reduced. Furthermore, SU6656 interfered with Aurora kinase activity resulting in inhibition of cell division and formation multilobular nuclei after 24h. Comparable alterations were observed in primary tubular cells. When cell division was inhibited by SU6656 or ZM447439, a specific inhibitor of Aurora kinases, CTGF levels were back to control or even increased after 48h. The activity of RhoA-Rho kinase and ERK signaling was analyzed to delineate the signaling pathways responsible for the biphasic regulation of CTGF. While Rho kinase was not significantly altered by SU6656, ERK activity was inhibited in the early phase and increased after 24-48h. ERK activity correlated with secreted CTGF. As ZM447439 increased ERK activity only after 48h, cellular reorganization is likely responsible for triggering the ERK-dependent upregulation of CTGF. Taken together, in non-transformed epithelial cells, SU6656 modulates the expression of the pro-fibrotic factor CTGF in a time-dependent manner by inhibition of Src kinases and Aurora kinases.


Subject(s)
Aurora Kinases/antagonists & inhibitors , Connective Tissue Growth Factor/biosynthesis , Indoles/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , src-Family Kinases/antagonists & inhibitors , Aurora Kinases/metabolism , Cell Adhesion/drug effects , Cell Culture Techniques , Cell Division/drug effects , Connective Tissue Growth Factor/genetics , Enzyme Activation/drug effects , Humans , Immunohistochemistry , Kidney Tubules/cytology , Kidney Tubules/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects , rho-Associated Kinases/metabolism , src-Family Kinases/metabolism
17.
Cell Commun Signal ; 11: 80, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24144209

ABSTRACT

BACKGROUND: Hypoxia is a major driving force in vascularization and vascular remodeling. Pharmacological inhibition of prolyl hydroxylases (PHDs) leads to an oxygen-independent and long-lasting activation of hypoxia-inducible factors (HIFs). Whereas effects of HIF-stabilization on transcriptional responses have been thoroughly investigated in endothelial cells, the molecular details of cytoskeletal changes elicited by PHD-inhibition remain largely unknown. To investigate this important aspect of PHD-inhibition, we used a spheroid-on-matrix cell culture model. RESULTS: Microvascular endothelial cells (glEND.2) were organized into spheroids. Migration of cells from the spheroids was quantified and analyzed by immunocytochemistry. The PHD inhibitor dimethyloxalyl glycine (DMOG) induced F-actin stress fiber formation in migrating cells, but only weakly affected microvascular endothelial cells firmly attached in a monolayer. Compared to control spheroids, the residual spheroids were larger upon PHD inhibition and contained more cells with tight VE-cadherin positive cell-cell contacts. Morphological alterations were dependent on stabilization of HIF-1α and not HIF-2α as shown in cells with stable knockdown of HIF-α isoforms. DMOG-treated endothelial cells exhibited a reduction of immunoreactive Rac-1 at the migrating front, concomitant with a diminished Rac-1 activity, whereas total Rac-1 protein remained unchanged. Two chemically distinct Rac-1 inhibitors mimicked the effects of DMOG in terms of F-actin fiber formation and orientation, as well as stabilization of residual spheroids. Furthermore, phosphorylation of p21-activated kinase PAK downstream of Rac-1 was reduced by DMOG in a HIF-1α-dependent manner. Stabilization of cell-cell contacts associated with decreased Rac-1 activity was also confirmed in human umbilical vein endothelial cells. CONCLUSIONS: Our data demonstrates that PHD inhibition induces HIF-1α-dependent cytoskeletal remodeling in endothelial cells, which is mediated essentially by a reduction in Rac-1 signaling.


Subject(s)
Actin Cytoskeleton/metabolism , Endothelial Cells/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Microvessels/drug effects , Prolyl-Hydroxylase Inhibitors/pharmacology , rac1 GTP-Binding Protein/metabolism , Actin Cytoskeleton/ultrastructure , Actins/metabolism , Amino Acids, Dicarboxylic/pharmacology , Cell Movement , Endothelial Cells/physiology , Endothelial Cells/ultrastructure , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Microvessels/physiology , Microvessels/ultrastructure , Signal Transduction , Spheroids, Cellular/cytology , Spheroids, Cellular/physiology , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/genetics , rho-Associated Kinases/metabolism
18.
Cell Commun Signal ; 10(1): 25, 2012 Sep 02.
Article in English | MEDLINE | ID: mdl-22938209

ABSTRACT

BACKGROUND: Increased expression of the pro-fibrotic protein connective tissue growth factor (CTGF) has been detected in injured kidneys and elevated urinary levels of CTGF are discussed as prognostic marker of chronic kidney disease. There is evidence that epithelial cells lining the renal tubular system contribute to uptake and secretion of CTGF. However, the role of different types of tubular epithelial cells in these processes so far has not been addressed in primary cultures of human cells. RESULTS: Tubular epithelial cells of proximal and distal origin were isolated from human kidneys and cultured as polarized cells in insert wells. The pro-fibrotic stimuli lysophosphatidic acid (LPA) and transforming growth factor ß (TGF-ß) were used to induce CTGF secretion.LPA activated CTGF secretion in proximal tubular cells when applied from either the apical or the basolateral side as shown by immunocytochemistry. CTGF was secreted exclusively to the apical side. Signaling pathways activated by LPA included MAP kinase and Rho kinase signaling. TGF-ß applied from either side also stimulated CTGF secretion primarily to the apical side with little basolateral release.Interestingly, TGF-ß activation induced different signaling pathways depending on the side of TGF-ß application. Smad signaling was almost exclusively activated from the basolateral side most prominently in cells of distal origin. Only part of these cells also synthesized CTGF indicating that Smad activation alone was not sufficient for CTGF induction. MAP kinases were involved in apical TGF-ß-mediated activation of CTGF synthesis in proximal cells and a subset of epithelial cells of distal origin. This subpopulation of distal tubular cells was also able to internalize recombinant apical CTGF, in addition to proximal cells which were the main cells to take up exogenous CTGF. CONCLUSIONS: Analysis of polarized human primary renal epithelial cells in a transwell system shows that vectorial secretion of the pro-fibrotic protein CTGF depends on the cell type, the stimulus and the signaling pathway activated. In all conditions, CTGF was secreted mainly to the apical side upon TGF-ß and LPA treatment and therefore, likely contributes to increased urinary CTGF levels in vivo. Moreover, CTGF secreted basolaterally may be active as paracrine pro-fibrotic mediator.

19.
PLoS One ; 7(8): e43584, 2012.
Article in English | MEDLINE | ID: mdl-22912891

ABSTRACT

BACKGROUND: Renal tubular epithelial cells of proximal and distal origin differ markedly in their physiological functions. Therefore, we hypothesized that they also differ in their capacity to undergo epithelial to mesenchymal alterations. RESULTS: We used cultures of freshly isolated primary human tubular cells. To distinguish cells of different tubular origin we took advantage of the fact that human proximal epithelial cells uniquely express N-cadherin instead of E-cadherin as major cell-cell adhesion molecule. To provoke mesenchymal alteration we treated these cocultures with TGF-ß for up to 6 days. Within this time period, the morphology of distal tubular cells was barely altered. In contrast to tubular cell lines, E-cadherin was not down-regulated by TGF-ß, even though TGF-ß signal transduction was initiated as demonstrated by nuclear localization of Smad2/3. Analysis of transcription factors and miRNAs possibly involved in E-cadherin regulation revealed high levels of miRNAs of the miR200-family, which may contribute to the stability of E-cadherin expression in human distal tubular epithelial cells. By contrast, proximal tubular epithelial cells altered their phenotype when treated with TGF-ß. They became elongated and formed three-dimensional structures. Rho-kinases were identified as modulators of TGF-ß-induced morphological alterations. Non-specific inhibition of Rho-kinases resulted in stabilization of the epithelial phenotype, while partial effects were observed upon downregulation of Rho-kinase isoforms ROCK1 and ROCK2. The distinct reactivity of proximal and distal cells was retained when the cells were cultured as polarized cells. CONCLUSIONS: Interference with Rho-kinase signaling provides a target to counteract TGF-ß-mediated mesenchymal alterations of epithelial cells, particularly in proximal tubular epithelial cells. Furthermore, primary distal tubular cells differed from cell lines by their high phenotypic stability which included constant expression of E-cadherin. Our cell culture system of primary epithelial cells is thus suitable to understand and modulate cellular remodeling processes of distinct tubular cells relevant for human renal disease.


Subject(s)
Cadherins/metabolism , Epithelial Cells/metabolism , Mesoderm/metabolism , Blotting, Western , Cadherins/genetics , Cell Adhesion/drug effects , Cell Polarity/drug effects , Cells, Cultured , Coculture Techniques , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kidney Tubules, Distal/cytology , Kidney Tubules, Proximal/cytology , Mesoderm/cytology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Zinc Finger E-box-Binding Homeobox 1 , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
20.
BMC Nephrol ; 13: 27, 2012 May 14.
Article in English | MEDLINE | ID: mdl-22583611

ABSTRACT

BACKGROUND: Microdeletions on chromosome 17q12 cause of diverse spectrum of disorders and have only recently been identified as a rare cause of Mayer-Rokitansky-Kuester-Hauser-Syndrome (MRKH), which is characterized by uterus aplasia ± partial/complete vaginal aplasia in females with a regular karyotype. For the first time we report about a patient with a 17q12 microdeletion who is affected by MRKH in combination with a vascular and soft tissue disorder. Repeatedly she suffered from kidney transplant failure caused by consuming membranous nephropathy. CASE PRESENTATION: A 38-year-old female patient had been diagnosed with right kidney aplasia, left kidney dysplasia and significantly impaired renal function during infancy. Aged 16 she had to start hemodialysis. Three years later she received her first kidney transplant. Only then she was diagnosed with MRKH. The kidney transplant was lost due to consuming nephrotic syndrome caused by de novo membranous nephropathy, as was a second kidney transplant years later. In addition, a hyperelasticity syndrome affects the patient with congenital joint laxity, kyphoscoliosis, bilateral hip dysplasia, persistent hypermobility of both elbows, knees and hips. Her clinical picture resembles a combination of traits of a hypermobile and a vascular form of Ehlers-Danlos-Syndrome, but no mutations in the COL3A1 gene was underlying. Instead, array-based comparative genomic hybridisation (CGH) detected a heterozygous 1.43 Mb deletion on chromosome 17q12 encompassing the two renal developmental genes HNF1ß and LHX1. CONCLUSIONS: Deletions of HNF1ß have recently drawn significant attention in pediatric nephrology as an important cause of prenatally hyperechogenic kidneys, renal aplasia and renal hypodysplasia. In contrast, membranous nephropathy represents an often-unaccounted cause of nephrotic syndrome in the adult population. A causative connection between theses two conditions has never been postulated, but is suggestive enough in this case to hypothesize it.


Subject(s)
Abnormalities, Multiple/genetics , Glomerulonephritis, Membranous/genetics , Phenotype , 46, XX Disorders of Sex Development , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/therapy , Adult , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Congenital Abnormalities , Female , Glomerulonephritis, Membranous/diagnosis , Glomerulonephritis, Membranous/therapy , Humans , Kidney/abnormalities , Mullerian Ducts/abnormalities , Recurrence , Smith-Magenis Syndrome , Somites/abnormalities , Spine/abnormalities , Uterus/abnormalities , Vagina/abnormalities
SELECTION OF CITATIONS
SEARCH DETAIL
...