Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
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
2.
Kidney Int ; 94(5): 887-899, 2018 11.
Article in English | MEDLINE | ID: mdl-30173898

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is mainly caused by mutations of the PKD1 gene and characterized by growth of bilateral renal cysts. Cyst growth is accompanied by regional hypoxia and induction of hypoxia-inducible factor (HIF)-1α in cyst-lining epithelial cells. To determine the relevance of HIF-1α for cyst growth in vivo we used an inducible kidney epithelium-specific knockout mouse to delete Pkd1 at postnatal day 20 or 35 to induce polycystic kidney disease of different severity and analyzed the effects of Hif-1α co-deletion and HIF-1α stabilization using a prolyl-hydroxylase inhibitor. HIF-1α expression was enhanced in kidneys with progressive cyst growth induced by early Pkd1 deletion, but unchanged in the milder phenotype induced by later Pkd1 deletion. Hif-1α co-deletion significantly attenuated cyst growth in the severe, but not in the mild, phenotype. Application of a prolyl-hydroxylase inhibitor resulted in severe aggravation of the mild phenotype with rapid loss of renal function. HIF-1α expression was associated with induction of genes that mediate calcium-activated chloride secretion. Thus, HIF-1α does not seem to play a role in early cyst formation, but accelerates cyst growth during progressive polycystic kidney disease. This novel mechanism of cyst growth may qualify as a therapeutic target.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Polycystic Kidney, Autosomal Dominant/etiology , Animals , Disease Models, Animal , Disease Progression , Mice , Polycystic Kidney, Autosomal Dominant/therapy
3.
Kidney Blood Press Res ; 43(1): 1-11, 2018.
Article in English | MEDLINE | ID: mdl-29393223

ABSTRACT

BACKGROUND/AIMS: One potential pathomechanism how low nephron number leads to hypertension in later life is altered salt handling. We therefore evaluated changes in electrolyte and water content in wildtype (wt) and GDNF+/- mice with a 30% reduction of nephron number. METHODS: 32 GDNF+/- and 36 wt mice were fed with low salt (LSD, 0.03%, normal drinking water) or high salt (HSD, 4%, 0.9% drinking water) diet for 4 weeks. Blood pressure was continuously measured by telemetry in a subgroup. At the end of the experiment and after standardized ashing processes electrolyte- and water contents of the skin and the total body were determined. RESULTS: We found higher blood pressure in high salt treated GDNF+/-compared to wt mice. Of interest, we could not confirm an increase in total-body sodium as predicted by prevailing explanations, but found increased total body and skin chloride that interestingly correlated with relative kidney weight. CONCLUSION: We hereby firstly report significant total body and skin chloride retention in salt sensitive hypertension of GDNF+/-mice with genetically determined lower nephron number. Thus, in contrast to the prevailing opinion our data argue for the involvement of non-volume related mechanisms.


Subject(s)
Chlorides/metabolism , Hypertension/etiology , Nephrons , Animals , Chlorides/analysis , Glial Cell Line-Derived Neurotrophic Factor/genetics , Kidney/physiology , Mice , Organ Size , Sodium/analysis , Sodium Chloride, Dietary
4.
Kidney Int ; 91(3): 616-627, 2017 03.
Article in English | MEDLINE | ID: mdl-27927598

ABSTRACT

Chronic kidney disease (CKD) is associated with increased risk and worse prognosis of cardiovascular disease, including peripheral artery disease. An impaired angiogenic response to ischemia may contribute to poor outcomes of peripheral artery disease in patients with CKD. Hypoxia inducible factors (HIF) are master regulators of angiogenesis and therefore represent a promising target for therapeutic intervention. To test this we induced hind-limb ischemia in rats with CKD caused by 5/6 nephrectomy and administered two different treatments known to stabilize HIF protein in vivo: carbon monoxide and a pharmacological inhibitor of prolyl hydroxylation 2-(1-chloro-4- hydroxyisoquinoline-3-carboxamido) acetate (ICA). Expression levels of pro-angiogenic HIF target genes (Vegf, Vegf-r1, Vegf-r2, Ho-1) were measured by qRT-PCR. Capillary density was measured by CD31 immunofluorescence staining and HIF expression was evaluated by immunohistochemistry. Capillary density in ischemic skeletal muscle was significantly lower in CKD animals compared to sham controls. Rats with CKD showed significantly lower expression of HIF and all measured pro-angiogenic HIF target genes, including VEGF. Both HIF stabilizing treatments rescued HIF target gene expression in animals with CKD and led to significantly higher ischemia-induced capillary sprouting compared to untreated controls. ICA was effective regardless of whether it was administered before or after induction of ischemia and led to a HIF expression in skeletal muscle. Thus, impaired ischemia-induced angiogenesis in rats with CKD can be improved by HIF stabilization, even if started after onset of ischemia.


Subject(s)
Capillaries/drug effects , Carbon Monoxide/pharmacology , Glycine/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/drug therapy , Isoquinolines/pharmacology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Renal Insufficiency, Chronic/metabolism , Signal Transduction/drug effects , Animals , Capillaries/metabolism , Capillaries/physiopathology , Cell Line , Disease Models, Animal , Gene Expression Regulation , Glycine/pharmacology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Hindlimb , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Male , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein Stability , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Kidney Int ; 88(6): 1283-1292, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26200943

ABSTRACT

Reduced nephron number predisposes to hypertension and kidney disease. Interaction of the branching ureteric bud and surrounding mesenchymal cells determines nephron number. Since oxygen supply may be critical for intrauterine development, we tested whether hypoxia and hypoxia-inducible factor-1α (HIF-1α) influence nephrogenesis. We found that HIF-1α is required for branching of MDCK cells. In addition, culture of metanephric mouse kidneys with ureteric bud cell-specific stabilization or knockout of HIF-1α revealed a positive impact of HIF-1α on nephrogenesis. In contrast, widespread stabilization of HIF-1α in metanephric kidneys through hypoxia or HIF stabilizers impaired nephrogenesis, and pharmacological HIF inhibition enhanced nephrogenesis. Several lines of evidence suggest an inhibitory effect through the hypoxia response of mesenchymal cells. HIF-1α was expressed in mesenchymal cells during nephrogenesis. Expression of the anti-branching factors Bmp4 and Vegfa, secreted by mesenchymal cells, was increased upon HIF stabilization. The conditioned medium from hypoxic metanephric kidneys inhibited MDCK branching, which was partially rescued by Vegfa antibodies. Thus, the effect of HIF-1α on nephrogenesis appears context dependent. While HIF-1α in the ureteric bud is of importance for proper branching morphogenesis, the net effect of hypoxia-induced HIF activation in the embryonic kidney appears to be mesenchymal cell-dependent inhibition of ureter branching.

6.
J Mol Med (Berl) ; 93(8): 891-904, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25754172

ABSTRACT

UNLABELLED: Hypoxia-inducible transcription factors (HIFs) control cellular adaptation to low oxygen. In the kidney, activation of HIF is beneficial during injury; however, the specific contribution of HIF-1α in renal endothelial cells (EC) remains elusive. Since EC display tissue-specific heterogeneity, we investigated how HIF-1α affects key functions of glomerular EC in vitro and its contribution to renal development and pathophysiological adaptation to acute or chronic renal injury in vivo. Loss of HIF-1α in glomerular EC induces hypoxic cell death and reduces hypoxic adhesion of macrophages in vitro. In vivo, HIF-1α expression in EC in mouse kidneys is detectable but limited. Accordingly, EC-specific ablation of HIF-1α does not lead to developmental or phenotypical abnormalities in the kidney. Renal function and expression of adhesion molecules during acute ischemic kidney injury is independent of HIF-1α in EC. Likewise, inflammation and development of fibrosis after unilateral ureteric obstruction is not influenced by endothelial HIF-1α. Taken together, although HIF-1α exerts effects on glomerular EC in vitro, endothelial HIF-1α does not influence renal development and pathophysiological adaptation to kidney injury in vivo. This implies a profound difference of the hypoxic response of the renal vascular bed compared to other organs, such as the heart. This has implications for the development of pharmacological strategies targeting the endothelial hypoxic response pathways. KEY MESSAGE: HIF-1α controls hypoxic survival and adhesion on endothelial cells (EC) in vitro. In vivo, HIF-1α expression in renal EC is low. Deletion of HIF-1α in EC does not affect kidney development and function in mice. Renal function after acute and chronic kidney injury is independent of HIF-1α in EC. Data suggest organ-specific regulation of HIF-1α function in EC.


Subject(s)
Endothelial Cells/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/injuries , Kidney/pathology , Reperfusion Injury/pathology , Animals , Cell Line , Endothelial Cells/metabolism , Fibrosis , Gene Deletion , Gene Expression Regulation, Developmental , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney/growth & development , Kidney/metabolism , Mice , Reperfusion Injury/genetics , Reperfusion Injury/metabolism
7.
PLoS One ; 9(2): e88601, 2014.
Article in English | MEDLINE | ID: mdl-24586350

ABSTRACT

BACKGROUND: Cardiovascular morbidity and mortality is very important in patients with chronic renal failure. This occurs even in mild impairment of renal function and may be related to oxidative stress and chronic inflammation. The nephrectomized apo E knockout mouse is an accepted model for evaluating atherosclerosis in renal dysfunction. Erythropoietin derivates showed anti-oxidative and anti-inflammatory effects. Therefore, this study evaluates the effects of Darbepoetin on markers of oxidative stress and chronic inflammation in atherosclerotic lesions in apo E knockout mice with renal dysfunction. METHODS: Apo E knockout mice underwent unilateral (Unx, n = 20) or subtotal (Snx, n = 26) nephrectomy or sham operation (Sham, n = 16). Mice of each group were either treated with Darbepoetin or saline solution, a part of Snx mice received a tenfold higher dose of Darbepoetin. The aortic plaques were measured and morphologically characterized. Additional immunhistochemical analyses were performed on tissue samples taken from the heart and the aorta. RESULTS: Both Unx and Snx mice showed increased expression of markers of oxidative stress and chronic inflammation. While aortic plaque size was not different, Snx mice showed advanced plaque stages when compared to Unx mice. Darbepoetin treatment elevated hematocrit and lowered Nitrotyrosin as one marker of oxidative stress, inflammation in heart and aorta, plaque stage and in the high dose even plaque cholesterol content. In contrast, there was no influence of Darbepoetin on aortic plaque size; high dose Darbepoetin treatment resulted in elevated renal serum parameters. CONCLUSION: Darbepoetin showed some protective cardiovascular effects irrespective of renal function, i.e. it improved plaque structure and reduced some signs of oxidative stress and chronic inflammation without affecting plaque size. Nevertheless, the dose dependent adverse effects must be considered as high Darbepoetin treatment elevated serum urea. Elevation of hematocrit might be a favorable effect in anemic Snx animals but a thrombogenic risk in Sham animals.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Erythropoietin/analogs & derivatives , Inflammation/drug therapy , Inflammation/metabolism , Renal Insufficiency/drug therapy , Renal Insufficiency/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Darbepoetin alfa , Erythropoietin/therapeutic use , Inflammation/genetics , Mice , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/genetics , Renal Insufficiency/genetics
8.
Biochim Biophys Acta ; 1842(4): 558-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24418215

ABSTRACT

Diabetes can disrupt endoplasmic reticulum (ER) homeostasis which leads to ER stress. ER stress-induced renal apoptosis seems to be involved in the development of diabetic nephropathy. The present study was designed to investigate the contribution of reduced ER stress to the beneficial effects of an angiotensin receptor blocker. Insulin-dependent diabetes mellitus was induced by streptozotocin injections to hypertensive mRen2-transgenic rats. After 2weeks animals were treated with 0.7mg/kg/day irbesartan. Blood glucose, blood pressure and protein excretion were assessed. Expression of ER stress markers was measured by real-time PCR. Immunohistochemistry was performed to detect markers of ER stress, renal damage and infiltrating cells. Glomerulosclerosis and apoptosis were evaluated. Diabetic mRen2-transgenic rats developed renal injury with proteinuria, tubulointerstitial cell proliferation as well as glomerulosclerosis and podocyte injury. Moreover, an increase in inflammation, podocyte ER stress and apoptosis was detected. Irbesartan somewhat lowered blood pressure and reduced proteinuria, tubulointerstitial cell proliferation and glomerulosclerosis. Podocyte damage was ameliorated but markers of ER stress (calnexin, grp78) and apoptosis were not reduced by irbesartan. On the other hand, inflammatory cell infiltration in the tubulointerstitium and the glomerulus was significantly attenuated. We conclude that irbesartan reduced renal damage even in a very low dose. The beneficial effects of low dose irbesartan were paralleled by a reduction of blood pressure and inflammation but not by a reduction of ER stress and apoptosis. Thus, sustained endoplasmic reticulum stress in the kidney does not necessarily lead to increased inflammation and tubulointerstitial or glomerular injury.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Biphenyl Compounds/therapeutic use , Diabetic Nephropathies/drug therapy , Endoplasmic Reticulum Stress/drug effects , Inflammation/prevention & control , Kidney/drug effects , Tetrazoles/therapeutic use , Animals , Apoptosis/drug effects , Blood Glucose/analysis , Blood Pressure/drug effects , Irbesartan , Male , Rats , Smad Proteins/physiology , Transforming Growth Factor beta/physiology
9.
J Am Soc Nephrol ; 25(3): 465-74, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24203996

ABSTRACT

Polycystic kidney diseases are characterized by numerous bilateral renal cysts that continuously enlarge and, through compression of intact nephrons, lead to a decline in kidney function over time. We previously showed that cyst enlargement is accompanied by regional hypoxia, which results in the stabilization of hypoxia-inducible transcription factor-1α (HIF-1α) in the cyst epithelium. Here we demonstrate a correlation between cyst size and the expression of the HIF-1α-target gene, glucose transporter 1, and report that HIF-1α promotes renal cyst growth in two in vitro cyst models-principal-like MDCK cells (plMDCKs) within a collagen matrix and cultured embryonic mouse kidneys stimulated with forskolin. In both models, augmenting HIF-1α levels with the prolyl hydroxylase inhibitor 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate enhanced cyst growth. In addition, inhibition of HIF-1α degradation through tubule-specific knockdown of the von Hippel-Lindau tumor suppressor increased cyst size in the embryonic kidney cyst model. In contrast, inhibition of HIF-1α by chetomin and knockdown of HIF-1α both decreased cyst growth in these models. Consistent with previous reports, plMDCK cyst enlargement was driven largely by transepithelial chloride secretion, which consists, in part, of a calcium-activated chloride conductance. plMDCKs deficient for HIF-1α almost completely lacked calcium-activated chloride secretion. We conclude that regional hypoxia in renal cysts contributes to cyst growth, primarily due to HIF-1α-dependent calcium-activated chloride secretion. These findings identify the HIF system as a novel target for inhibition of cyst growth.


Subject(s)
Chlorides/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Polycystic Kidney Diseases/etiology , Animals , Chloride Channels/metabolism , Dogs , Female , Gene Expression Regulation , Glucose Transport Proteins, Facilitative/metabolism , Hypoxia/physiopathology , Madin Darby Canine Kidney Cells , Male , Mice, Inbred C57BL , Polycystic Kidney Diseases/metabolism
10.
Mol Carcinog ; 53(12): 970-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23818324

ABSTRACT

Hypoxia leads to the upregulation of a variety of genes mediated largely via the hypoxia inducible transcription factor (HIF). Prominent HIF-regulated target genes such as the vascular endothelial growth factor (VEGF), the glucose transporter 1 (Glut-1), or erythropoietin (EPO) help to assure survival of cells and organisms in a low oxygenated environment. Here, we are the first to report the hypoxic regulation of the sperm associated antigen 4 (SPAG4). SPAG4 is a member of the cancer testis (CT) gene family and to date little is known about its physiological function or its involvement in tumor biology. A number of CT family candidate genes are therefore currently being investigated as potential cancer markers, due to their predominant testicular expression pattern. We analyzed RNA and protein expression by RNAse protection assay, immunofluorescent as well as immunohistological stainings. To evaluate the influence of SPAG4 on migration and invasion capabilities, siRNA knockdown as well as transient overexpression was performed prior to scratch or invasion assay analysis. The hypoxic regulation of SPAG4 is clearly mediated in a HIF-1 and VHL dependent manner. We furthermore show upregulation of SPAG4 expression in human renal clear cell carcinoma (RCC) and co-localization within the nucleolus in physiological human testis tissue. SPAG4 knockdown reduces the invasion capability of RCC cells in vitro and overexpression leads to enhancement of tumor cell migration. Together, SPAG4 could possibly play a role in the invasion capability and growth of renal tumors and could represent an interesting target for clinical intervention.


Subject(s)
Carcinoma, Renal Cell/genetics , Carrier Proteins/genetics , Cell Movement/genetics , Hypoxia-Inducible Factor 1/genetics , Hypoxia/genetics , Kidney Neoplasms/genetics , Neoplasm Invasiveness/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , HeLa Cells , Humans , Up-Regulation/genetics
11.
PLoS One ; 7(11): e48362, 2012.
Article in English | MEDLINE | ID: mdl-23144868

ABSTRACT

In the normal kidney, the α8 integrin chain is expressed only on mesangial cells and vascular smooth muscle cells. α8 integrin ligates several matrix molecules including fibronectin, osteopontin and fibrillin-1. Recently, we detected de novo expression of α8 integrin on epithelial cells in renal cysts. We hypothesized that the α8 integrin chain is induced in tubular epithelia undergoing dedifferentiation and contributes to the fibrotic response in the tubulointerstitium (TI) after unilateral ureteral obstruction (UUO). After induction of UUO in rats by ligation of the right ureter, increased expression of the α8 integrin chain and its ligands was observed. In the TI, α8 integrin was localized to cytokeratin-positive epithelial cells and to interstitial fibroblasts; and colocalized with its ligands. In mice underexpressing α8 integrin UUO led to collagen deposition and fibroblast activation comparable to wild types. Mice lacking α8 integrin showed even more TI damage, fibroblast activation and collagen deposition after UUO compared to wild type mice. We conclude that the expression of the α8 integrin chain and its ligands is strongly induced in the TI after UUO, but underexpression of α8 integrin does not attenuate TI fibrosis. Mice lacking the α8 integrin chain are even more susceptible to TI damage than wild type mice. Thus, interactions of α8 integrin with its ligands do not seem to contribute to the development or progression of TI fibrosis in UUO. Targeting α8 integrin might not be a useful approach for anti-fibrotic therapy.


Subject(s)
Integrin alpha Chains/metabolism , Kidney Diseases/metabolism , Kidney Tubules/metabolism , Actins/metabolism , Animals , Collagen Type I/metabolism , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Fibrillin-1 , Fibrillins , Fibroblasts/metabolism , Fibronectins/metabolism , Fibrosis , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Kidney Tubules/pathology , Male , Mice , Mice, Knockout , Microfilament Proteins/metabolism , Molecular Targeted Therapy , Osteopontin/metabolism , Protein Transport , Rats , Rats, Sprague-Dawley , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
12.
Am J Pathol ; 181(5): 1595-606, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22944601

ABSTRACT

The role of proximal versus distal tubular injury in the pathogenesis of acute kidney injury (AKI) is debatable. Inhibition of prolyl hydroxylases that regulate the degradation of hypoxia-inducible transcription factors (HIFs) is a promising therapeutic approach to optimize energy preservation under hypoxia and has successfully been applied to protect kidney structure and function in AKI models. Presently used prolyl hydroxylase inhibitors are lipophilic 2-oxoglutarate analogues (2OGAs) that are widely taken up in cells of most organs. Given the selective expression of organic anion transporters (OATs) in renal proximal tubular cells, we hypothesized that hydrophilic 2OGAs can specifically target proximal tubular cells. We found that cellular hydrophilic 2OGAs uptake depended on OATs and largely confined to the kidney, where it resulted in activation of HIF target genes only in proximal tubular cells. When applied in ischemia-reperfusion experiments, systemically active 2OGA preserved kidney structure and function, but OAT1-transported 2OGA was not protective, suggesting that HIF stabilization in distal tubular rather than proximal tubular cells and/or nontubular cells mediates protective effects. This study provides proof of concept for selective drug targeting of proximal tubular cells on the basis of specific transporters, gives insights into the role of different nephron segments in AKI pathophysiology, and may offer options for long-term HIF stabilization in proximal tubules without confounding effects of erythropoietin induction in peritubular cells and unwarranted extrarenal effects.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ketoglutaric Acids/chemistry , Ketoglutaric Acids/pharmacology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Acetic Acid/chemistry , Acetic Acid/pharmacology , Acute Kidney Injury/complications , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Amino Acids, Dicarboxylic/pharmacology , Animals , Biological Transport/drug effects , Cell Line , Cell Separation , Gene Expression Regulation/drug effects , Humans , Ischemia/complications , Ischemia/pathology , Ischemia/physiopathology , Ischemic Preconditioning , Kidney Function Tests , Kidney Tubules, Proximal/drug effects , Mice , Organic Anion Transport Protein 1/metabolism , Protein Stability/drug effects , Pyridines/chemistry , Pyridines/pharmacology
13.
PLoS One ; 7(1): e31034, 2012.
Article in English | MEDLINE | ID: mdl-22299048

ABSTRACT

The Hypoxia-inducible transcription Factor (HIF) represents an important adaptive mechanism under hypoxia, whereas sustained activation may also have deleterious effects. HIF activity is determined by the oxygen regulated α-subunits HIF-1α or HIF-2α. Both are regulated by oxygen dependent degradation, which is controlled by the tumor suppressor "von Hippel-Lindau" (VHL), the gatekeeper of renal tubular growth control. HIF appears to play a particular role for the kidney, where renal EPO production, organ preservation from ischemia-reperfusion injury and renal tumorigenesis are prominent examples. Whereas HIF-1α is inducible in physiological renal mouse, rat and human tubular epithelia, HIF-2α is never detected in these cells, in any species. In contrast, distinct early lesions of biallelic VHL inactivation in kidneys of the hereditary VHL syndrome show strong HIF-2α expression. Furthermore, knockout of VHL in the mouse tubular apparatus enables HIF-2α expression. Continuous transgenic expression of HIF-2α by the Ksp-Cadherin promotor leads to renal fibrosis and insufficiency, next to multiple renal cysts. In conclusion, VHL appears to specifically repress HIF-2α in renal epithelia. Unphysiological expression of HIF-2α in tubular epithelia has deleterious effects. Our data are compatible with dedifferentiation of renal epithelial cells by sustained HIF-2α expression. However, HIF-2α overexpression alone is insufficient to induce tumors. Thus, our data bear implications for renal tumorigenesis, epithelial differentiation and renal repair mechanisms.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression , Kidney Diseases, Cystic/genetics , Kidney Tubules/metabolism , Kidney Tubules/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , COS Cells , Chlorocebus aethiops , Fibrosis/genetics , Gene Expression/physiology , Gene Silencing/physiology , HEK293 Cells , HeLa Cells , Humans , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Kidney Tubules/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Opossums , Rats , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
14.
J Am Soc Nephrol ; 22(11): 2004-15, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21921145

ABSTRACT

Hypoxia-inducible transcription factors (HIF) protect cells against oxygen deprivation, and HIF stabilization before ischemia mitigates tissue injury. Because ischemic acute kidney injury (AKI) often involves the thick ascending limb (TAL), modulation of HIF in this segment may be protective. Here, we generated mice with targeted TAL deletion of the von Hippel-Lindau protein (Vhl), which mediates HIF degradation under normoxia, using Tamm-Horsfall protein (Thp)-driven Cre expression. These mice showed strong expression of HIF-1α in TALs but no changes in kidney morphology or function under control conditions. Deficiency of Vhl in the TAL markedly attenuated proximal tubular injury and preserved TAL function following ischemia-reperfusion, which may be partially a result of enhanced expression of glycolytic enzymes and lactate metabolism. These results highlight the importance of the thick ascending limb in the pathogenesis of AKI and suggest that pharmacologically targeting the HIF system may have potential to prevent and mitigate AKI.


Subject(s)
Acute Kidney Injury/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Loop of Henle/physiology , Uromodulin/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Anaerobic Threshold/physiology , Animals , Disease Models, Animal , Erythropoiesis/physiology , Glycolysis/physiology , Integrases/genetics , Kidney/physiology , Mice , Mice, Mutant Strains , Nephritis/genetics , Nephritis/metabolism , Nephritis/physiopathology , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Uromodulin/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
15.
Nephrol Dial Transplant ; 26(11): 3458-65, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21804086

ABSTRACT

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of renal failure. Aberrant epithelial cell proliferation is a major cause of progressive cyst enlargement in ADPKD. Since activation of the Ras/Raf signaling system has been detected in cyst-lining epithelia, inhibition of Raf kinase has been proposed as an approach to retard the progression of ADPKD. Methods and results. PLX5568, a novel selective small molecule inhibitor of Raf kinases, attenuated proliferation of human ADPKD cyst epithelial cells. It reduced in vitro cyst growth of Madin-Darby Canine Kidney cells and of human ADPKD cells within a collagen gel. In male cy/+ rats with polycystic kidneys, PLX5568 inhibited renal cyst growth along with a significant reduction in the number of proliferating cell nuclear antigen- and phosphorylated extracellular signal-regulated kinase-positive cyst-lining epithelial cells. Furthermore, treated animals showed increased capacity to concentrate urine. However, PLX5568 did not lead to a consistent improvement of renal function. Moreover, although relative cyst volume was decreased, total kidney-to-body weight ratio was not significantly reduced by PLX5568. Further analyses revealed a 2-fold increase of renal and hepatic fibrosis in animals treated with PLX5568. CONCLUSIONS: PLX5568 attenuated cyst enlargement in vitro and in a rat model of ADPKD without improving kidney function, presumably due to increased renal fibrosis. These data suggest that effective therapies for the treatment of ADPKD will need to target fibrosis as well as the growth of cysts.


Subject(s)
Cell Proliferation/drug effects , Cysts/pathology , Kidney/physiopathology , Liver Cirrhosis/chemically induced , Polycystic Kidney Diseases/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Cells, Cultured , Cysts/drug therapy , Dogs , Epithelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases , Humans , Immunoblotting , Immunoenzyme Techniques , Kidney/drug effects , Male , Mitogen-Activated Protein Kinases , Phosphorylation/drug effects , Polycystic Kidney Diseases/enzymology , Polycystic Kidney Diseases/pathology , Protein Kinase Inhibitors/toxicity , Rats , Rats, Sprague-Dawley
16.
J Hypertens ; 29(8): 1602-12, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21610512

ABSTRACT

BACKGROUND: The plasminogen-plasmin system affects tissue fibrosis, presumably by interacting with metalloproteinases (MMPs) and macrophage recruitment. This study tests the influence of plasminogen activator inhibitor-1 (PAI-1) and tissue-type plasminogen activator (tPa) on angiotensin II-mediated hypertensive kidney and heart injury. METHOD: Hypertension was induced by continuous angiotensin II (Ang II) infusion via osmotic mini-pumps over 4 weeks. The effects of Ang II infusion were determined in mice lacking PAI-1 (PAI-1), mice lacking tPa (tPa), and wild-type mice. Normotensive mice of the respective genotype served as controls. Blood pressure was recorded by continuous radiotelemetric intra-arterial measurements. RESULTS: Ang II infusion significantly enhanced arterial blood pressure in all groups. However, the increase in blood pressure was more pronounced in the tPa group. Albuminuria was highest in hypertensive wild-type compared to the other Ang II-infused groups. Hypertensive PAI-1 mice exhibited less glomerulosclerosis, higher nephrin immunostaining, and lower renal interstitial collagen I deposition. Gelatin zymography revealed higher activity of MMP-2 in hypertensive PAI-1, whereas no differences were observed in macrophage infiltration. tPa deficiency did not alter kidney fibrosis, although hypertensive tPa revealed less renal expression of fibrotic genes, less macrophage infiltration, and reduced MMP-2 activity. On the other hand, hypertension-induced fibrosis as well as macrophage infiltration in the heart was profoundly enhanced in PAI-1 mice. Fibrin staining revealed perivascular exudations in the myocardium of hypertensive PAI-1 suggesting vascular leakage. CONCLUSION: These findings underscore the unexpectedly complex role of plasminogen activation for hypertensive target organ damage.


Subject(s)
Fibrinolysin/physiology , Glomerulonephritis/epidemiology , Heart Diseases/epidemiology , Hypertension/complications , Hypertension/physiopathology , Plasminogen/physiology , Aldosterone/blood , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Disease Models, Animal , Fibrosis , Glomerulonephritis/physiopathology , Heart Diseases/physiopathology , Hypertension/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Plasminogen Activator Inhibitor 1/deficiency , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/physiology , Prevalence , Risk Factors , Tissue Plasminogen Activator/deficiency , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/physiology
17.
Biochim Biophys Acta ; 1813(1): 1-13, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21056597

ABSTRACT

Hypoxia is a common pathogenic stress, which requires adaptive activation of the Hypoxia-inducible transcription factor (HIF). In concert transcriptional HIF targets enhance oxygen availability and simultaneously reduce oxygen demand, enabling survival in a hypoxic microenvironment. Here, we describe the characterization of a new HIF-1 target gene, Rab20, which is a member of the Rab family of small GTP-binding proteins, regulating intracellular trafficking and vesicle formation. Rab20 is directly regulated by HIF-1, resulting in rapid upregulation of Rab20 mRNA as well as protein under hypoxia. Furthermore, exogenous as well as endogenous Rab20 protein colocalizes with mitochondria. Knockdown studies reveal that Rab20 is involved in hypoxia induced apoptosis. Since mitochondria play a key role in the control of cell death, we suggest that regulating mitochondrial homeostasis in hypoxia is a key function of Rab20. Furthermore, our study implicates that cellular transport pathways play a role in oxygen homeostasis. Hypoxia-induced Rab20 may influence tissue homeostasis and repair during and after hypoxic stress.


Subject(s)
Apoptosis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/pathology , Mitochondria/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Caspases/metabolism , Cells, Cultured , Electrophoretic Mobility Shift Assay , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/genetics
18.
Kidney Int ; 78(9): 857-67, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20720525

ABSTRACT

The two hypoxia-inducible factors (HIF-1α and HIF-2α) are transcription factors that regulate the response to hypoxia. Recently, the factor inhibiting HIF (FIH1) was identified as a molecular oxygen-dependent dioxygenase that blunts the transcriptional activity of HIF and has also been implicated in HIF-dependent and -independent hypoxia responses. Interestingly, HIF accumulation in the kidney has been shown to confer renal protection and to also cause glomerular injury or enhance renal fibrosis. In order to better understand the regulation of hypoxia-inducible genes, we determined the expression of FIH1 in the kidney and its functional role in isolated renal cells. FIH1 was expressed only in distal tubules and in podocytes, thus showing a very distinct expression pattern, partially overlapping with sites of HIF-1α expression. In tubular cells, RNA silencing of FIH1 caused transcriptional activation of HIF target genes during hypoxia. In contrast, FIH1 silencing in podocytes enhanced transcription of hypoxia-inducible genes in an HIF-independent manner. Using the anti-Thy.1 rat model of glomerulonephritis, we found a gradual decrease of glomerular FIH1 expression during disease progression paralleled by an increase in hypoxia-inducible genes including CXCR4, a mediator of glomerular inflammation. Thus, FIH1 appears to be a suppressor of oxygen-dependent genes in the kidney, operating through HIF-dependent and -independent mechanisms.


Subject(s)
Glomerulonephritis, Membranoproliferative/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Tubules, Distal/metabolism , Mixed Function Oxygenases/metabolism , Oxygen/metabolism , Podocytes/metabolism , Repressor Proteins/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Isoantibodies , Male , Mice , Mice, Inbred C57BL , Mixed Function Oxygenases/genetics , RNA Interference , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Time Factors
19.
PLoS One ; 4(11): e7875, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19924283

ABSTRACT

BACKGROUND: Hepcidin is a major regulator of iron metabolism and plays a key role in anemia of chronic disease, reducing intestinal iron uptake and release from body iron stores. Hypoxia and chemical stabilizers of the hypoxia-inducible transcription factor (HIF) have been shown to suppress hepcidin expression. We therefore investigated the role of HIF in hepcidin regulation. METHODOLOGY/PRINCIPAL FINDINGS: Hepcidin mRNA was down-regulated in hepatoma cells by chemical HIF stabilizers and iron chelators, respectively. In contrast, the response to hypoxia was variable. The decrease in hepcidin mRNA was not reversed by HIF-1alpha or HIF-2alpha knock-down or by depletion of the HIF and iron regulatory protein (IRP) target transferrin receptor 1 (TfR1). However, the response of hepcidin to hypoxia and chemical HIF inducers paralleled the regulation of transferrin receptor 2 (TfR2), one of the genes critical to hepcidin expression. Hepcidin expression was also markedly and rapidly decreased by serum deprivation, independent of transferrin-bound iron, and by the phosphatidylinositol 3 (PI3) kinase inhibitor LY294002, indicating that growth factors are required for hepcidin expression in vitro. Hepcidin promoter constructs mirrored the response of mRNA levels to interleukin-6 and bone morphogenetic proteins, but not consistently to hypoxia or HIF stabilizers, and deletion of the putative HIF binding motifs did not alter the response to different hypoxic stimuli. In mice exposed to carbon monoxide, hypoxia or the chemical HIF inducer N-oxalylglycine, liver hepcidin 1 mRNA was elevated rather than decreased. CONCLUSIONS/SIGNIFICANCE: Taken together, these data indicate that hepcidin is neither a direct target of HIF, nor indirectly regulated by HIF through induction of TfR1 expression. Hepcidin mRNA expression in vitro is highly sensitive to the presence of serum factors and PI3 kinase inhibition and parallels TfR2 expression.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Hypoxia-Inducible Factor 1/metabolism , Iron-Regulatory Proteins/chemistry , Amino Acid Motifs , Animals , Antigens, CD/metabolism , Base Sequence , Chromones/pharmacology , Hepcidins , Humans , Interleukin-6/metabolism , Mice , Molecular Sequence Data , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Receptors, Transferrin/metabolism
20.
Am J Pathol ; 174(5): 1663-74, 2009 May.
Article in English | MEDLINE | ID: mdl-19349364

ABSTRACT

Hypoxia-inducible transcription factors (HIFs) play important roles in the response of the kidney to systemic and regional hypoxia. Degradation of HIFs is mediated by three oxygen-dependent HIF-prolyl hydroxylases (PHDs), which have partially overlapping characteristics. Although PHD inhibitors, which can induce HIFs in the presence of oxygen, are already in clinical development, little is known about the expression and regulation of these enzymes in the kidney. Therefore, we investigated the expression levels of the three PHDs in both isolated tubular cells and rat kidneys. All three PHDs were present in the kidney and were expressed predominantly in three different cell populations: (a) in distal convoluted tubules and collecting ducts (PHD1,2,3), (b) in glomerular podocytes (PHD1,3), and (c) in interstitial fibroblasts (PHD1,3). Higher levels of PHDs were found in tubular segments of the inner medulla where oxygen tensions are known to be physiologically low. PHD expression levels were unchanged in HIF-positive tubular and interstitial cells after induction by systemic hypoxia. In rat models of acute renal injury, changes in PHD expression levels were variable; while cisplatin and ischemia/reperfusion led to significant decreases in PHD2 and 3 expression levels, no changes were seen in a model of contrast media-induced nephropathy. These results implicate the non-uniform expression of HIF-regulating enzymes that modify the hypoxic response in the kidney under both regional and temporal conditions.


Subject(s)
Acute Kidney Injury/enzymology , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immediate-Early Proteins/metabolism , Kidney/enzymology , Procollagen-Proline Dioxygenase/metabolism , Animals , Antineoplastic Agents/toxicity , Blotting, Western , Cisplatin/toxicity , Contrast Media/pharmacology , DNA-Binding Proteins/genetics , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Regulation, Enzymologic/physiology , Homeodomain Proteins/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases , Immediate-Early Proteins/genetics , Immunoenzyme Techniques , Ischemia/metabolism , Ischemia/pathology , Kidney/drug effects , Kidney/injuries , Kidney Medulla/drug effects , Kidney Medulla/enzymology , Kidney Tubules/drug effects , Kidney Tubules/enzymology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/enzymology , Male , Mice , Mice, Inbred C57BL , Oxygen/metabolism , Podocytes/drug effects , Podocytes/enzymology , Procollagen-Proline Dioxygenase/genetics , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...