Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Physiol (Oxf) ; 224(3): e13102, 2018 11.
Article in English | MEDLINE | ID: mdl-29791781

ABSTRACT

AIM: In acute kidney injury (AKI), regions of the kidney are hypoxic. However, for reasons yet unknown, adaptation to hypoxia through hypoxia-inducible factor (HIF) is limited. Here, we studied miR-22, a potential HIF repressor, in normal kidneys, as well as in rhabdomyolysis-induced AKI, a condition where miR-22 is up-regulated. METHODS: AKI in mice was provoked by IM injection of glycerol. Tissue homogenates were processed to determine the levels of candidate RNAs and proteins, as well as global gene expression profiles. Reporter assays quantified in vitro miR-22 activity and its modulation by mimic or inhibitor molecules, under normoxia or hypoxia (1% O2 ) respectively. In vivo, anti-miR-22 molecules were applied to normal mice or prior to induction of AKI. Renal outcome was assessed by measuring plasma creatinine, plasma urea and the levels of the injury markers Kim-1 and Ngal. RESULTS: Renal miR-22 is inducible by hypoxia and represses hypoxia-inducible factor (HIF). Specific inhibition of miR-22 regulates 1913 gene transcripts in kidneys controls and 3386 in AKI, many of which are involved in development or carcinogenesis. Specific inhibition of miR-22 up-regulates tissue protective HIF target genes, yet renal function and injury markers are unchanged or worsened. CONCLUSIONS: miR-22 is a HIF repressor constitutively expressed in the adult kidney and up-regulated in AKI. Specific inhibition of miR-22 is efficient in vivo and profoundly affects renal gene expression in health and disease, including up-regulation of HIF. However, the net effect on rhabdomyolysis-induced AKI outcome is neutral or even negative.


Subject(s)
Acute Kidney Injury/metabolism , MicroRNAs/metabolism , Rhabdomyolysis/metabolism , Animals , Gene Expression Regulation , Glycerol/administration & dosage , Glycerol/toxicity , Kidney Tubules, Distal/drug effects , Kidney Tubules, Distal/metabolism , Kidney Tubules, Distal/pathology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Solvents/administration & dosage , Solvents/toxicity
2.
Acta Physiol (Oxf) ; 223(1): e13033, 2018 05.
Article in English | MEDLINE | ID: mdl-29330945

ABSTRACT

AIM: Cyclosporine A (CsA) induces renal vasoconstriction and hypoxia and enhances the expression of endothelin-1 (ET-1) pro-hormone (pre-pro-ET-1), plausibly leading to a feed-forward loop of renal vasoconstriction, hypoxia and enhanced synthesis of the potent vasoconstrictor ET-1. Endothelin-converting enzyme (ECE)-1 cleaves big endothelin to generate endothelin (ET)-1 and is upregulated by hypoxia via hypoxia-inducible factor (HIF). We hypothesized that in addition to the direct induction of ET-1 synthesis, CsA might also intensify renal ECE-1 expression, thus contributing to enhanced ET-1 synthesis following CsA. METHODS: CsA was administered to Sprague Dawley rats (120 mg/kg/SC) for 4 days, and renal HIF and ECE-1 expression were assessed with Western blots and immunostaining. Human umbilical vein endothelial cells (HUVEC) and proximal tubular cell line (HK-2) were subjected to CsA, and ECE-1 induction was evaluated using real-time mRNA PCR and Western blots. RESULTS: Cyclosporine A intensified renal parenchymal ECE-1 expression in the rat kidney, particularly in distal nephron segments, along with renal hypoxia (detected by pimonidazole adducts) and HIF expression, in line with our recent observations showing episodic hypoxia in mice subjected to CsA. Furthermore, in cultured normoxic HUVEC and HK-2 cells, CsA dose-dependently induced both pre-pro-ET-1 and ECE-1 mRNA and protein expression, with enhanced ET-1 generation. CONCLUSION: CsA induces ECE-1 via both hypoxic and non-hypoxic pathways. ECE-1 may contribute to increased renal ET-1 generation following CsA, participating in a feed-forward loop of renal parenchymal hypoxia and ET synthesis.


Subject(s)
Cyclosporine/pharmacology , Endothelin-Converting Enzymes/biosynthesis , Human Umbilical Vein Endothelial Cells/drug effects , Kidney/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Endothelin-1/genetics , Endothelin-1/metabolism , Endothelin-Converting Enzymes/blood , Endothelin-Converting Enzymes/genetics , Enzyme Induction , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/enzymology , Male , Rats, Sprague-Dawley , Up-Regulation
3.
Am J Physiol Renal Physiol ; 312(5): F908-F916, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28052871

ABSTRACT

Vasoconstriction plays an important role in the development of acute kidney injury in rhabdomyolysis. We hypothesized that myoglobin enhances the angiotensin II (ANG II) response in afferent arterioles by increasing superoxide and reducing nitric oxide (NO) bioavailability. Afferent arterioles of C57Bl6 mice were isolated perfused, and vasoreactivity was analyzed using video microscopy. NO bioavailability, superoxide concentration in the vessel wall, and changes in cytosolic calcium were measured using fluorescence techniques. Myoglobin treatment (10-5 M) did not change the basal arteriolar diameter during a 20-min period compared with control conditions. NG-nitro-l-arginine methyl ester (l-NAME, 10-4 M) and l-NAME + myoglobin reduced diameters to 94.7 and 97.9% of the initial diameter, respectively. Myoglobin or l-NAME enhanced the ANG II-induced constriction of arterioles compared with control (36.6 and 34.2%, respectively, vs. 65.9%). Norepinephrine responses were not influenced by myoglobin. Combined application of myoglobin and l-NAME further facilitated the ANG II response (7.0%). Myoglobin or l-NAME decreased the NO-related fluorescence in arterioles similarly. Myoglobin enhanced the superoxide-related fluorescence, and tempol prevented this enhancement. Tempol also partly prevented the myoglobin effect on the ANG II response. Myoglobin increased the fura 2 fluorescence ratio (cytosolic calcium) during ANG II application (10-12 to 10-6 M). The results suggest that the enhanced afferent arteriolar reactivity to ANG II is mainly due to a myoglobin-induced increase in superoxide and associated reduction in the NO bioavailability. Signaling pathways for the augmented ANG II response include enhanced cytosolic calcium transients. In conclusion, myoglobin may contribute to the afferent arteriolar vasoconstriction in this rhabdomyolysis model.


Subject(s)
Angiotensin II/pharmacology , Arterioles/drug effects , Kidney/blood supply , Myoglobin/pharmacology , Rhabdomyolysis/physiopathology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Antioxidants/pharmacology , Arterioles/metabolism , Arterioles/physiopathology , Calcium Signaling/drug effects , Cyclic N-Oxides/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Male , Mice, Inbred C57BL , Microscopy, Video , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Oxidative Stress/drug effects , Rhabdomyolysis/metabolism , Spin Labels , Superoxides/metabolism , Time Factors
4.
Acta Physiol (Oxf) ; 219(3): 625-639, 2017 03.
Article in English | MEDLINE | ID: mdl-27690155

ABSTRACT

AIM: Cyclosporin A (CsA) causes renal toxicity. The underlying mechanisms are incompletely understood, but may involve renal hypoxia and hypoxia-inducible factors (Hifs). We sought for hypoxia and Hif in mouse kidneys with CsA-induced toxicity, assessed their time course, Hif-mediated responses and the impact of interventional Hif upregulation. METHODS: Mice received CsA or its solvent cremophore for up to 6 weeks. Low salt diet (Na+ ↓) was given in combination with CsA to enhance toxicity. We assessed fine morphology, renal function, blood oxygen level-dependent magnetic resonance imaging under room air and following changes in breathing gas composition which correlate with vascular reactivity, pimonidazole adducts (which indicate O2 tensions below 10 mmHg), Hif-α proteins, as well as expression of Hif target genes. Stable Hif upregulation was achieved by inducible, Pax8-rtTA-based knockout of von Hippel-Lindau protein (Vhl-KO), which is crucial for Hif-α degradation. RESULTS: Cyclosporin A transiently increased renal deoxyhaemoglobin (R2*). Augmented vascular reactivity was observed at 2 h, but decreased at 24 h after CsA treatment. Na+ ↓/CsA provoked chronic renal failure with tubular degeneration and interstitial fibrosis. Nephron segments at risk for injury accumulated pimonidazole adducts, as well as Hif-α proteins. Remarkably, Hif target gene expression remained unchanged, while factor-inhibiting Hif (Fih) was enhanced. Na+ ↓/CsA/Vhl-KO aggravated morpho-functional outcome of chronic renal CsA toxicity. CONCLUSIONS: Cyclosporin A provokes episodic hypoxia in nephron segments most susceptible to chronic CsA toxicity. Fih is upregulated and likely blocks further Hif activity. Continuous tubular Hif upregulation via Vhl-KO worsens the outcome of chronic CsA-induced renal toxicity.


Subject(s)
Cell Hypoxia/drug effects , Cyclosporine/toxicity , Immunosuppressive Agents/toxicity , Kidney Diseases/chemically induced , Kidney/drug effects , Animals , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/metabolism , Kidney Diseases/metabolism , Mice , Mice, Knockout , Mixed Function Oxygenases/metabolism , Up-Regulation
5.
Acta Physiol (Oxf) ; 207(3): 565-76, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23384425

ABSTRACT

AIM: Von Hippel-Lindau protein (VHL) provides the degradation of hypoxia-inducible factor (HIF). Tetracycline-induced, Pax8-rtTA-based knockout of VHL (VHL-KO) affects all renal tubules and periportal hepatocytes and leads to sustained upregulation of HIF. Here, we study the phenotype of VHL-KO in both organs, the time course of changes, and long-term morpho-functional outcome. METHODS: Mice with doxycycline-induced VHL-KO and controls (CON) were followed for up to 9 months. Systemic and tissue parameters were evaluated using clinical chemistry, histology, immunohistochemistry, RT-PCR and in situ hybridisation. RESULTS: At day 3 following VHL-KO, substantial abundance of HIF-1α and -2α was detected in the nuclei of hepatocytes and renal tubular epithelia. Hypoxia, induced by bleeding anaemia, did not further augment HIF signal. Erythropoietin mRNA was detectable in hepatocytes but not in the kidney. Vascular endothelial growth factor mRNA was upregulated in kidney but not in liver. At day 7 following VHL-KO, the renal capillary density was enhanced, reaching its maximum at day 14. Blood haemoglobin increased constantly up to day 28 (23.3 vs. 15.8 g dL(-1) , VHL-KO vs. CON). Thereafter, it was kept within the normal range by weekly blood collections. Pathological changes were absent from kidney and liver 9 months after VHL-KO. CONCLUSIONS: Inducible, Pax8-rtTA-based deletion of VHL leads to organ-specific expression of epithelial HIF and erythropoietin in liver and kidney without causing pathological changes. Uniform, maximal and sustained HIF activation along the renal tubule may serve to study the potential benefits of hypoxia adaptation in experimental renal injury.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/metabolism , Liver/metabolism , Paired Box Transcription Factors/genetics , Trans-Activators/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Capillaries/metabolism , Erythropoiesis , Erythropoietin/genetics , Erythropoietin/metabolism , Female , Genotype , Hemoglobins/metabolism , Immunohistochemistry , In Situ Hybridization , Kidney/blood supply , Male , Mice , Mice, Knockout , Mice, Transgenic , Neovascularization, Physiologic , PAX8 Transcription Factor , Phenotype , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Time Factors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics
6.
Acta Physiol (Oxf) ; 207(4): 721-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23347696

ABSTRACT

AIM: In the present study, we aimed to elucidate the effects of chronic vasopressin administration on renal medullary oxygen levels. METHODS: Adult Sprague Dawley or vasopressin-deficient Brattleboro rats were treated with the vasopressin V2 receptor agonist, desmopressin (5 ng/h; 3d), or its vehicle via osmotic minipumps. Immunostaining for pimonidazole and the transcription factor HIF-1α (hypoxia-inducible factor-1α) were used to identify hypoxic areas. Activation of HIF-target gene expression following desmopressin treatment was studied by microarray analysis. RESULTS: Pimonidazole staining was detected in the outer and inner medulla of desmopressin-treated rats, whereas staining in control animals was weak or absent. HIF-1α immunostaining demonstrated nuclear accumulation in the papilla of desmopressin-treated animals, whereas no staining was observed in the controls. Gene expression analysis revealed significant enrichment of HIF-target genes in the group of desmopressin-regulated gene products (P = 2.6*10(-21) ). Regulated products included insulin-like growth factor binding proteins 1 and 3, angiopoietin 2, fibronectin, cathepsin D, hexokinase 2 and cyclooxygenase 2. CONCLUSION: Our results demonstrate that an activation of the renal urine concentrating mechanism by desmopressin causes renal medullary hypoxia and an upregulation of hypoxia-inducible gene expression.


Subject(s)
Hypoxia/metabolism , Kidney Medulla/metabolism , Oxygen/metabolism , Receptors, Vasopressin/physiology , Signal Transduction/physiology , Animals , Deamino Arginine Vasopressin/pharmacology , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Medulla/drug effects , Nitroimidazoles/metabolism , Rats , Rats, Brattleboro , Rats, Sprague-Dawley , Receptors, Vasopressin/agonists , Receptors, Vasopressin/drug effects , Vasopressins/deficiency , Vasopressins/genetics , Vasopressins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...