Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Int J Mol Sci ; 25(1)2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38203251

ABSTRACT

Proximal tubular epithelial cells (PTEC) are constantly exposed to potentially toxic metabolites and xenobiotics. The regenerative potential of the kidney enables the replacement of damaged cells either via the differentiation of stem cells or the re-acquisition of proliferative properties of the PTEC. Nevertheless, it is known that renal function declines, suggesting that the deteriorated cells are not replaced by fully functional cells. To understand the possible causes of this loss of kidney cell function, it is crucial to understand the role of toxins during the regeneration process. Therefore, we investigated the sensitivity and function of human induced pluripotent stem cells (hiPSC), hiPSC differentiating, and hiPSC differentiated into proximal tubular epithelial-like cells (PTELC) to known nephrotoxins. hiPSC were differentiated into PTELC, which exhibited similar morphology to PTEC, expressed prototypical PTEC markers, and were able to undergo albumin endocytosis. When treated with two nephrotoxins, hiPSC and differentiating hiPSC were more sensitive to cisplatin than differentiated PTELC, whereas all stages were equally sensitive to cyclosporin A. Both toxins also had an inhibitory effect on albumin uptake. Our results suggest a high sensitivity of differentiating cells towards toxins, which could have an unfavorable effect on regenerative processes. To study this, our model of hiPSC differentiating into PTELC appears suitable.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Cell Differentiation , Kidney , Albumins , Epithelial Cells
2.
Int J Mol Sci ; 21(13)2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32630085

ABSTRACT

Hypertensive patients have an increased risk of developing chronic kidney disease (CKD). Many of these patients have increased levels of the blood pressure regulating mineralocorticoid aldosterone. As a protection against aldosterone-induced damage, kidney cells can upregulate key regulators of the antioxidant defense, such as nuclear factor-erythroid-2-related factor 2 (Nrf2). In the present study aldosterone-induced kidney damage and Nrf2 activation in kidney cells of mice treated with three different concentrations of aldosterone for 4 weeks was localized. Increased albumin and neutrophil gelatinase-associated lipocalin (NGAL) in urine revealed an impaired kidney function of the aldosterone-infused mice. Localization of aldosterone-induced oxidative damage (in the form of DNA lesions) in specific kidney cells showed an increase in proximal tubuli and to an even greater extend in distal tubuli. Phosphorylated Nrf2 was increased in distal tubule cells after aldosterone-infusion. Nrf2 activation in proximal tubuli or in glomeruli after aldosterone-treatment could not be observed. Nrf2 target genes and proteins analyzed, paradoxically, showed a downregulation in the whole kidney. Aldosterone-treated mice exhibited an increased kidney injury and DNA damage in distal and proximal tubuli. Nrf2 seemed only to be specifically activated in distal tubule cells, where we also detected the highest amount of oxidative damage.


Subject(s)
Aldosterone/blood , Kidney/metabolism , NF-E2-Related Factor 2/metabolism , Renal Insufficiency, Chronic/blood , Aldosterone/administration & dosage , Animals , Blood Pressure , DNA Damage , Kidney/pathology , Male , Mice, Inbred C57BL , Renal Insufficiency, Chronic/pathology
3.
Biochim Biophys Acta Mol Cell Res ; 1865(9): 1277-1292, 2018 09.
Article in English | MEDLINE | ID: mdl-29932987

ABSTRACT

BACKGROUND: The Ras-homologous GTPase Rac1 plays a key role in the regulation of gene expression, cytoskeleton-associated processes and cell death as well as carcinogenesis and inflammation. Here, we investigated the impact of Rac1 signaling on liver-mediated immune homeostasis. METHODS: We employed a constitutive Alb-Cre-driven rac1 knock-out and a poly I:C-inducible Mx1-Cre-based knock-out model and analyzed cytokine expression profiles in liver and other organs under basal situation and following LPS-induced endotoxemia by flow cytometry, qRT-PCR and immunocytochemistry. RESULTS: Constitutive Alb-Cre-driven rac1 knockout in hepatocytes altered the basal distribution and activation of immune cells in the liver and likewise in kidney and lung. Early systemic alterations in cytokine serum levels following LPS treatment remained unaffected by Rac1. Furthermore, lack of Rac1 in hepatocytes of untreated animals shifted the liver to a chronic inflammatory state, as depicted by an enhanced mRNA expression of marker genes related to activated macrophages. Upon acute LPS-induced endotoxemia, increased IL-10 mRNA expression in the liver of Alb-Cre Rac1-deficient mice provided an anti-inflammatory response. Employing a poly I:C-inducible Mx1-Cre-based rac1 knock-out, which allows a more widespread rac1 deletion in both hepatocytes and non-hepatocytes, we observed substantial differences regarding both basal and LPS-stimulated cytokine expression profiles as compared to the Alb-Cre system. CONCLUSIONS: Rac1-dependent mechanisms in hepatocytes and non-hepatocytes contribute to the maintenance of liver immune homeostasis under basal situation and following LPS-induced endotoxemia. Disturbed Rac1-regulated hepatocyte functions may promote liver damage under pathophysiological situation involving inflammatory stress.


Subject(s)
Endotoxemia/enzymology , Interleukin-10/genetics , Lipopolysaccharides/adverse effects , Liver/immunology , Neuropeptides/genetics , Neuropeptides/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , Animals , Disease Models, Animal , Endotoxemia/chemically induced , Endotoxemia/genetics , Endotoxemia/immunology , Gene Expression Regulation , Gene Knockout Techniques , Immunity , Kidney/immunology , Liver/enzymology , Lung/immunology , Macrophages/metabolism , Mice , Signal Transduction
4.
Cell Physiol Biochem ; 52(1): 27-39, 2019.
Article in English | MEDLINE | ID: mdl-30790503

ABSTRACT

BACKGROUND/AIMS: SGLT-2 inhibitors have been shown to be nephroprotective in diabetes. Here, we examined if one of these drugs (canagliflozin) could also ameliorate non-diabetic chronic kidney disease (CKD). METHODS: CKD was induced in rats by feeding them adenine (0.25%w/w for 35 days) and canagliflozin (10 or 25 mg/kg, by gavage) was given with or without adenine. Several conventional and novel plasma and urine biomarkers and tissues morphology were used to investigate the canagliflozin effect on kidney structure and function. RESULTS: Rats fed adenine showed the typical features of CKD that included elevation of blood pressure, decreased food intake and growth, increased water intake and urine output, decrease in creatinine clearance, and increase in urinary albumin/creatinine ratio, liver-type fatty acid binding protein, N-acetyl-beta-D-glucosaminidase, and plasma urea, creatinine, uric acid, calcium, indoxyl sulfate and phosphorus concentrations. Adenine also increased concentrations of several biomarkers of inflammation such as neutrophil gelatinase-associated lipocalin, interleukin-6, tumor necrosis factor alpha, clusterin, cystatin C and interleukin-1ß, and decreased some oxidative biomarkers in kidney homogenate, such as superoxide dismutase, catalase, glutathione reductase, total antioxidant activity, and also urinary 8-isoprostane and urinary 8-hydroxy-2-deoxy guanosine. Adenine significantly increased the renal protein content of Nrf2, caused renal tubular necrosis and fibrosis. Given alone, canagliflozin at the two doses used did not significantly alter any of the parameters mentioned above. When canagliflozin was given concomitantly with adenine, it significantly and dose - dependently ameliorated all the measured adenine - induced actions. CONCLUSION: Canagliflozin ameliorated adenine - induced CKD in rats, through reduction of several inflammatory and oxidative stress parameters, and other indices such as uremic toxins, and by antagonizing the increase in the renal content of the transcription factor Nrf2. The drug caused no overt or significant untoward effects, and its trial in patients with CKD may be warranted.


Subject(s)
Adenine/adverse effects , Canagliflozin/pharmacology , Renal Insufficiency, Chronic , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Adenine/pharmacology , Animals , Biomarkers/urine , Rats , Rats, Wistar , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/prevention & control , Renal Insufficiency, Chronic/urine
5.
Arch Toxicol ; 93(7): 2045-2055, 2019 07.
Article in English | MEDLINE | ID: mdl-31098697

ABSTRACT

Epidemiological studies revealed an increased risk for kidney cancer in hypertensive patients. In many of these patients, the blood pressure regulating renin-angiotensin-aldosterone system (RAAS) is activated. A stimulated RAAS leads to oxidative stress and increases markers of DNA damage, both in vitro and in animal models of hypertension. However, the mutagenic potential of RAAS activation has not been investigated yet. To quantify hypertension-induced mutations, BigBlue®+/- rats, which carry a transgenic lacI gene for mutation analysis, were treated for 20 weeks with a mean dose of 400 µg angiotensin II/kg × day. Angiotensin II-treated animals showed significantly increased blood pressure and impaired kidney function. Urinary excretion of oxidized nucleobases was raised. Additionally, in the renal cortex, oxidative stress, oxidatively generated DNA lesions and DNA strandbreaks were significantly increased. Further, a significant elevation of the mutant frequency in kidney DNA was detected. Sequencing revealed the presence of GC → T:A transversions in the mutated lacI genes of the angiotensin II-treated animals as a result of unrepaired oxidatively modified DNA bases, while no such transversions were found in the mutated lacI genes from control animals. The results demonstrate that the oxidative stress and DNA damage previously observed in kidney cells in vitro and in vivo after angiotensin II treatment indeed is associated with the accumulation of mutations in rat kidneys, providing further evidence for a cancer-initiating potential of elevated angiotensin II concentrations.


Subject(s)
Angiotensin II/metabolism , Hypertension/physiopathology , Kidney/physiopathology , Renin-Angiotensin System/physiology , Angiotensin II/administration & dosage , Animals , Blood Pressure/physiology , DNA Damage/physiology , Hypertension/complications , Lac Repressors/genetics , Male , Mutation , Oxidative Stress/physiology , Rats , Rats, Transgenic
6.
Am J Physiol Renal Physiol ; 313(6): F1264-F1273, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28877878

ABSTRACT

Hypertensive patients have an increased risk of developing kidney cancer. We have shown in vivo that besides elevating blood pressure, angiotensin II causes DNA damage dose dependently. Here, the role of blood pressure in the formation of DNA damage is studied. Mice lacking one of the two murine angiotensin II type 1 receptor (AT1R) subtypes, AT1aR, were equipped with osmotic minipumps, delivering angiotensin II during 28 days. Parameters of oxidative stress and DNA damage of kidneys and hearts of AT1aR-knockout mice were compared with wild-type (C57BL/6) mice receiving angiotensin II, and additionally, with wild-type mice treated with candesartan, an antagonist of both AT1R subtypes. In wild-type mice, angiotensin II induced hypertension, reduced kidney function, and led to a significant formation of reactive oxygen species (ROS). Furthermore, genomic damage was markedly increased in this group. All these responses to angiotensin II could be attenuated by concurrent administration of candesartan. In AT1aR-deficient mice treated with angiotensin II, systolic pressure was not increased, and renal function was not affected. However, angiotensin II still led to an increase of ROS in kidneys and hearts of these animals. Additionally, genomic damage in the form of double-strand breaks was significantly induced in kidneys of AT1aR-deficient mice. Our results show that angiotensin II induced ROS production and DNA damage even without the presence of AT1aR and independently of blood pressure changes.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Blood Pressure/drug effects , DNA Damage/drug effects , Oxidative Stress/drug effects , Receptor, Angiotensin, Type 1/deficiency , Angiotensin II/pharmacology , Animals , Hypertension/metabolism , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/metabolism
7.
Mol Carcinog ; 56(8): 1868-1883, 2017 08.
Article in English | MEDLINE | ID: mdl-28272757

ABSTRACT

Epidemiological studies found an increased risk for kidney cancer in hypertensive patients, of which a subgroup has high aldosterone (Ald) levels. We recently showed that Ald is genotoxic both in kidney tubular cells and in rats with mineralocorticoid-mediated hypertension. The present work investigated in vitro and in vivo, if the oxidative stress-mediated activation of the ERK1/2 pathway, and its downstream target STAT3, could be one mechanism involved in the potential oncogenic capability of excess Ald exposure. The effects of excess Ald were investigated in LLC-PK1 cells and in Ald-induced hypertensive rats. Ald caused cRaf, MEK1/2, and ERK1/2 phosphorylation both in LLC-PK1 cells and in rat kidneys. ERK1/2 activation led to an increased phosphorylation of MSK1, p90RSK, and STAT3. The involvement of ERK1/2 in the activation of STAT3 was evidenced by the capacity of the MEK inhibitor U0126 to prevent Ald-mediated ERK1/2 and STAT3 phosphorylation. Both in vitro and in vivo, the activation of ERK1/2 and STAT3 by Ald was dependent on the mineralocorticoid receptor and was triggered by an increase in cellular oxidants. Ald-mediated oxidant increase was in part due to the activation of the enzymes NADPH oxidase and NO synthase. Proliferation was significantly enhanced and apoptosis decreased in Ald-treated rat kidneys and/or LLC-PK1 cells. Results support the concept that the oxidant-mediated long-term activation of ERK1/2/STAT3 by persistently high Ald levels could trigger proliferative and prosurvival events. Ald-mediated promotion of cell survival and DNA damage could result in kidney cell transformation and initiation of cancer in hypertensive patients with hyperaldosteronism.


Subject(s)
Aldosterone/metabolism , Kidney Neoplasms/metabolism , Kidney/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxidative Stress , STAT3 Transcription Factor/metabolism , Animals , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic , Enzyme Activation , Humans , Kidney/pathology , Kidney/ultrastructure , Kidney Neoplasms/pathology , Male , NADPH Oxidases/metabolism , Nitric Oxide Synthase/metabolism , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
8.
Cell Physiol Biochem ; 38(1): 248-57, 2016.
Article in English | MEDLINE | ID: mdl-26784294

ABSTRACT

BACKGROUND/AIMS: To study the therapeutic effect of chrysin, a flavonoid with strong antioxidant and anti-inflammatory activities, on adenine-induced chronic kidney diseases (CKD) in rats. METHODS: Chrysin, in three graded oral doses (10, 50 and 250 mg/kg), was given for 10 consecutive days to rats after the induction of CKD by feeding them adenine (0.25%(w/w) for 35 days). Several plasma and urine biomarkers and tissues morphology were used the investigate chrysin effect on kidney structure and function. RESULTS: Adenine lowered creatinine clearance and elevated the concentrations of urea, creatinine, plasma neutrophil gelatinase-associated lipocalin and urinary N-Acetyl-beta-D-glucosaminidase activity, and increased the concentrations of the uremic toxin indoxyl sulfate, in addition to some inflammatory cytokines. Renal histopathological markers of inflammation and fibrosis were significantly increased. Renal catalase and superoxide dismutase activities, total antioxidant capacity and reduced glutathione were all adversely affected. Most of these adenine - induced actions were moderately mitigated by chrysin, especially at the highest dose. Compared to control, chrysin did not cause any overt adverse effects on the treated rats. CONCLUSION: Different doses of chrysin produce variable therapeutic salutary effects in rats with CKD, and that, pending further studies, its usability as a possible therapeutic agent in human CKD should be considered.


Subject(s)
Antioxidants/therapeutic use , Flavonoids/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Acute-Phase Proteins/urine , Adenine/toxicity , Administration, Oral , Animals , Antioxidants/chemistry , Biomarkers/blood , Biomarkers/urine , Catalase/metabolism , Creatinine/blood , Cytokines/blood , Disease Models, Animal , Flavonoids/chemistry , Glutathione/metabolism , Kidney/pathology , Lipocalin-2 , Lipocalins/urine , Proto-Oncogene Proteins/urine , Rats , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Superoxide Dismutase/metabolism , Urea/blood
9.
Toxicol Appl Pharmacol ; 292: 103-14, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26739623

ABSTRACT

The platinating agent cisplatin (CisPt) is commonly used in the therapy of various types of solid tumors. The anticancer efficacy of CisPt largely depends on the formation of bivalent DNA intrastrand crosslinks, which stimulate mechanisms of the DNA damage response (DDR), thereby triggering checkpoint activation, gene expression and cell death. The clinically most relevant adverse effect associated with CisPt treatment is nephrotoxicity that results from damage to renal tubular epithelial cells. Here, we addressed the question whether the HMG-CoA-reductase inhibitor lovastatin affects the DDR of renal cells by employing rat renal proximal tubular epithelial (NRK-52E) cells as in vitro model. The data show that lovastatin has extensive inhibitory effects on CisPt-stimulated DDR of NRK-52E cells as reflected on the levels of phosphorylated ATM, Chk1, Chk2, p53 and Kap1. Mitigation of CisPt-induced DDR by lovastatin was independent of the formation of DNA damage as demonstrated by (i) the analysis of Pt-(GpG) intrastrand crosslink formation by Southwestern blot analyses and (ii) the generation of DNA strand breaks as analyzed on the level of nuclear γH2AX foci and employing the alkaline comet assay. Lovastatin protected NRK-52E cells from the cytotoxicity of high CisPt doses as shown by measuring cell viability, cellular impedance and flow cytometry-based analyses of cell death. Importantly, the statin also reduced the level of kidney DNA damage and apoptosis triggered by CisPt treatment of mice. The data show that the lipid-lowering drug lovastatin extensively counteracts pro-apoptotic signal mechanisms of the DDR of tubular epithelial cells following CisPt injury.


Subject(s)
Apoptosis/drug effects , Cisplatin/toxicity , DNA Damage/drug effects , Epithelial Cells/drug effects , Kidney Tubules/drug effects , Lovastatin/pharmacology , Animals , Apoptosis/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cisplatin/antagonists & inhibitors , DNA Damage/physiology , Epithelial Cells/metabolism , Kidney Tubules/metabolism , Mice , Mice, Inbred BALB C , Rats
10.
Toxicol Appl Pharmacol ; 280(3): 399-407, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25204689

ABSTRACT

Mineralocorticoid receptor blockers show antifibrotic potential in hepatic fibrosis. The mechanism of this protective effect is not known yet, although reactive oxygen species seem to play an important role. Here, we investigated the effects of elevated levels of aldosterone (Ald), the primary ligand of the mineralocorticoid receptor, on livers of rats in a hyperaldosteronism model: aldosterone-induced hypertension. Male Sprague-Dawley rats were treated for 4 weeks with aldosterone. To distinguish if damage caused in the liver depended on increased blood pressure or on increased Ald levels, the mineralocorticoid receptor antagonist spironolactone was given in a subtherapeutic dose, not normalizing blood pressure. To investigate the impact of oxidative stress, the antioxidant tempol was administered. Aldosterone induced fibrosis, detected histopathologically, and by expression analysis of the fibrosis marker, α-smooth muscle actin. Further, the mRNA amount of the profibrotic cytokine TGF-ß was increased significantly. Fibrosis could be reduced by scavenging reactive oxygen species, and also by blocking the mineralocorticoid receptor. Furthermore, aldosterone treatment caused oxidative stress and DNA double strand breaks in livers, as well as the elevation of DNA repair activity. An increase of the transcription factor Nrf2, the main regulator of the antioxidative response could be observed, and of its target genes heme oxygenase-1 and γ-glutamylcysteine synthetase. All these effects of aldosterone were prevented by spironolactone and tempol. Already after 4 weeks of treatment, aldosteroneinfusion induced fibrosis in the liver. This effect was independent of elevated blood pressure. DNA damage caused by aldosterone might contribute to fibrosis progression when aldosterone is chronically increased.


Subject(s)
Aldosterone , Blood Pressure , Cyclic N-Oxides , Liver Cirrhosis , Mineralocorticoid Receptor Antagonists , Oxidative Stress , Spironolactone , Animals , Male , Aldosterone/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Blotting, Western , Cyclic N-Oxides/pharmacology , Histocytochemistry , Liver Cirrhosis/metabolism , Mineralocorticoid Receptor Antagonists/pharmacology , Oxidative Stress/physiology , Random Allocation , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , RNA/chemistry , RNA/genetics , Spin Labels , Spironolactone/pharmacology , Statistics, Nonparametric , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , NF-E2-Related Factor 2/metabolism
11.
Antioxidants (Basel) ; 12(3)2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36979025

ABSTRACT

Nuclear factor erythroid 2-related factor 2 (Nrf2) is downregulated in chronic kidney disease (CKD). Activation of Nrf2 might be a therapeutic option in CKD. Here we investigate the effect of Nrf2 activation on aldosterone (Aldo)-induced renal injury. Wild-type (WT) mice, transgenic Keap1 hypomorphic (Nrf2ꜛ, genotype results in upregulation of Nrf2 expression) mice and WT mice treated with the Nrf2 activator sulforaphane (Sulf) received Aldo for 4 weeks. In Aldo-treated mice, kidneys were significantly heavier and pathologically altered, reflected by increased urinary albumin levels and tissue damage. In Nrf2ꜛ-Aldo mice the tubule damage marker NGAL was significantly decreased. Increased oxidative damage markers (8-OHdG, 15-isoprostane F2t) were measured in all Aldo-treated groups. Aldo-increased Nrf2 amounts were mainly found in the late tubule system. The amount of phosphorylated and thus putatively active Nrf2 was significantly increased by Aldo only in WT mice. However, expression of Nrf2 target genes NQO1 and HO1 was decreased in all Aldo-infused mice. GSK3ß, which promotes Nrf2 degradation, was significantly increased in the kidneys of Aldo-treated WT mice. Neither genetic nor pharmacological Nrf2 activation was able to prevent oxidative injury induced by Aldo, probably due to induction of negative regulators of Nrf2.

12.
Antioxidants (Basel) ; 12(11)2023 Nov 04.
Article in English | MEDLINE | ID: mdl-38001818

ABSTRACT

Patients with arterial hypertension have an increased risk of developing tumors, particularly renal cell carcinoma. Arterial hypertension is linked to DNA damage via the generation of oxidative stress, in which an upregulated renin-angiotensin-aldosterone system plays a crucial role. The current study investigated surrogates of oxidative stress and DNA damage in a group of hypertensive patients (HypAll, n = 64) and subgroups of well (HypWell, n = 36) and poorly (HypPoor, n = 28) controlled hypertensive patients compared to healthy controls (n = 8). In addition, a longitudinal analysis was performed with some of the hypertensive patients. Markers for oxidative stress in plasma (SHp, D-ROM, and 3-nitrotyrosine) and urine (8-oxodG, 15-F2t-isoprostane, and malondialdehyde) and markers for DNA damage in lymphocytes (γ-H2AX and micronuclei) were measured. In HypAll, all markers of oxidative stress except malondialdehyde were increased compared to the controls. After adjustment for age, this association was maintained for the protein stress markers SHp and 3-nitrotyrosine. With regard to the markers for DNA damage, there was no difference between HypAll and the controls. Further, no significant differences became apparent in the levels of both oxidative stress and DNA damage between HypWell and HypPoor. Finally, a positive correlation between the development of blood pressure and oxidative stress was observed in the longitudinal study based on the changes in D-ROM and systolic blood pressure. In conclusion, we found increased oxidative stress in extensively treated hypertensive patients correlating with the level of blood-pressure control but no association with DNA damage.

13.
FASEB J ; 25(3): 968-78, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21135038

ABSTRACT

Epidemiological studies exploring the connection between hypertension and cancer demonstrate a higher cancer incidence, especially of kidney cancer, and a higher cancer mortality in hypertensive patients. Hormones elevated in hypertension, i.e., aldosterone and angiotensin II, which exert genotoxic effects in vitro, could contribute to carcinogenesis in hypertension. The present study was conducted to investigate the possible DNA-damaging effect of aldosterone receptor activation in vivo. Crl:CD (Sprague-Dawley) rats were treated for 6 wk with desoxycorticosterone acetate (DOCA) and salt to induce a mineralocorticoid-dependent hypertension. DOCA-salt treatment caused increased blood pressure (+26 mmHg) compared to untreated rats, elevated markers of kidney failure (up to 62-fold for Kim-1), and the induction of several proinflammatory genes and proteins (up to 2.6-fold for tissue MCP-1). The mineralocorticoid receptor (MR) antagonist spironolactone (MR IC(50) 24 nM) and the novel nonsteroidal antagonist BR-4628 (MR IC(50) 28 nM) decreased these damage markers. DOCA-salt treatment also caused 8.8-fold increased structural DNA damage, determined with the comet assay, double-strand breaks (3.5-fold), detected immunohistochemically, and oxidative stress. Furthermore, the oxidatively modified mutagenic DNA base 7,8-dihydro-8-oxo-guanine (8-oxodG), quantified by LC-MS/MS, was almost 2-fold higher in DOCA-salt-treated kidneys. Our results suggest a mutagenic potential of high mineralocorticoid levels, frequent in hypertensive individuals.


Subject(s)
DNA Breaks, Double-Stranded , DNA Damage/physiology , Hypertension, Renal/metabolism , Hypertension, Renal/pathology , Receptors, Mineralocorticoid/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blood Pressure/physiology , Cell Division/drug effects , Cell Division/physiology , Desoxycorticosterone/toxicity , Disease Models, Animal , Guanine/analogs & derivatives , Guanine/metabolism , Hypertension, Renal/drug therapy , Kidney/pathology , Kidney/physiopathology , Male , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoids/toxicity , Nephrectomy , Nephritis/drug therapy , Nephritis/metabolism , Nephritis/pathology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/genetics , Spironolactone/pharmacology
14.
Mutagenesis ; 27(6): 673-81, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22844079

ABSTRACT

Epidemiological studies revealed increased renal cancer incidences and higher cancer mortalities in hypertensive individuals. Activation of the renin-angiotensin-aldosterone system leads to the formation of reactive oxygen species (ROS). In vitro, in renal cells, and ex vivo, in the isolated perfused mouse kidney, we could show DNA-damaging potential of angiotensin II (Ang II). Here, the pathway involved in the genotoxicity of Ang II was investigated. In kidney cell lines with properties of proximal tubulus cells, an activation of NADPH oxidase and the production of ROS, resulting in the formation of DNA strand breaks and micronuclei induction, was observed. This DNA damage was mediated by the Ang II type 1 receptor (AT1R), together with the G protein G ( α-q/11 ) . Subsequently, phospholipase C (PLC) was activated and intracellular calcium increased. Both calcium stores of the endoplasmic reticulum and extracellular calcium were involved in the genotoxicity of Ang II. Downstream, a role for protein kinase C (PKC) could be detected, because its inhibition hindered Ang II from damaging the cells. Although PKC was activated, no involvement of its known target, the NADPH oxidase isoform containing the Nox2 subunit, could be found, as tested by small-interfering RNA down-regulation. Responsible for the DNA-damaging activity of Ang II was the NADPH oxidase isoform containing the Nox4 subunit. In summary, in kidney cells the DNA-damaging activity of Ang II depends on an AT1R-mediated activation of NADPH oxidase via PLC, PKC and calcium signalling, with the NADPH subunit Nox4 playing a crucial role.


Subject(s)
Angiotensin II/toxicity , DNA Damage/drug effects , NADPH Oxidases/metabolism , Receptor, Angiotensin, Type 1/metabolism , Animals , Calcium , Calcium Signaling , Cell Line , Down-Regulation , Kidney/cytology , Kidney/drug effects , Male , Mice , Mice, Inbred C57BL , NADPH Oxidase 4 , NADPH Oxidases/genetics , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/genetics , Type C Phospholipases/metabolism
15.
Mol Carcinog ; 50(2): 123-35, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21229609

ABSTRACT

An increase of the mineralocorticoid aldosterone is induced by a stimulated renin-angiotensin system in a subgroup of hypertensive patients. Epidemiological studies find higher cancer mortality in hypertensive patients and an increased risk to develop kidney cancer. This work investigated the involvement of oxidants in the genotoxicity of aldosterone and on a potential activation of transcription factor nuclear factor-κB (NF-κB) in kidney tubule cells. Aldosterone, at concentrations as low as 1 nM caused a significant increase of DNA damage, as assessed by comet assay and micronucleus frequency test. Aldosterone also led to a dose-dependent activation of NF-κB. Time courses of DNA damage and NF-κB-activation showed that these effects already occurred after 5 and 3 min of aldosterone exposure, respectively, suggesting non-genomic events of the hormone. Antioxidants prevented aldosterone-induced DNA damage and NF-κB-activation, indicating the involvement of oxidants. In fact, aldosterone caused an increase in intracellular oxidant levels, and in particular of superoxide anions. As a consequence, increased levels of the oxidized DNA modification 7,8-dihydro-8-oxo-guanine were observed in aldosterone-treated kidney cells. Aldosterone-induced DNA damage and NF-κB-activation was dependent on the involvement of the mineralocorticoid receptor. The induction of oxidant-mediated genotoxic effects, and of a long-term activation of the potentially oncogenic cell signal NF-κB by aldosterone could contribute to the increased kidney cancer incidence in hypertensive patients.


Subject(s)
Aldosterone/metabolism , DNA Damage , Kidney Neoplasms/metabolism , Kidney Tubules/pathology , NF-kappa B/metabolism , Oxidative Stress , Renin-Angiotensin System/physiology , Aldosterone/pharmacology , Animals , Antioxidants/pharmacology , Cell Line , Dogs , Guanine/analogs & derivatives , Guanine/analysis , Kidney Tubules/drug effects , Receptors, Mineralocorticoid/metabolism , Renin-Angiotensin System/drug effects , Superoxides/analysis , Swine
16.
Kidney Blood Press Res ; 34(3): 167-72, 2011.
Article in English | MEDLINE | ID: mdl-21474964

ABSTRACT

BACKGROUND: Angiotensin II (ANG II) and advanced glycation end products (AGEs) exert genotoxic effects in vitro which were prevented by the ANG II type 1 (AT1) receptor blocker, candesartan. In end-stage renal disease (ESRD) the incidence of genomic damage is increased. A stimulation of the renin-angiotensin system and accumulation of AGEs could be involved. METHODS: We tested whether oral co-administration of candesartan modulates enhanced DNA damage in ESRD patients. Fifteen maintenance hemodialysis (MHD) patients with mild hypertension were treated with candesartan for 4.5 months. Fourteen MHD patients served as conventionally treated uremic controls. DNA damage was measured as micronucleus frequency (MNF) in peripheral blood lymphocytes and evaluated three times before candesartan therapy and afterwards every 6 weeks. RESULTS: Compared to 14 healthy controls, MNF at baseline was significantly elevated in MHD patients. While in the conventionally treated MHD patients the enhanced DNA damage persisted, the co-administration of candesartan ameliorated the genomic damage significantly and independently of blood pressure changes. CONCLUSION: Blockade of AT1 receptors with candesartan can reduce DNA damage in MHD patients. Long-term studies in larger patient groups are needed to investigate whether the improved genomic damage lowers atherosclerotic complications and cancer development.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Antimutagenic Agents , Benzimidazoles/pharmacology , Lymphocytes/physiology , Lymphocytes/ultrastructure , Renal Dialysis/adverse effects , Tetrazoles/pharmacology , Adult , Aged , Aged, 80 and over , Angiotensin II/antagonists & inhibitors , Angiotensin II/toxicity , Antihypertensive Agents/therapeutic use , Biphenyl Compounds , Blood Pressure/physiology , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Separation , Female , Glycation End Products, Advanced/antagonists & inhibitors , Glycation End Products, Advanced/toxicity , Humans , Hypertension/drug therapy , Hypertension/etiology , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/therapy , Kidney Function Tests , Lymphocytes/drug effects , Male , Micronucleus Tests , Middle Aged
17.
Biol Chem ; 391(11): 1265-79, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20868230

ABSTRACT

The formation of reactive oxygen species (ROS) can be induced by xenobiotic substances, such as redox cycling molecules, but also by endogenous substances such as hormones and cytokines. Recent research shows the importance of ROS in cellular signaling. Here, the signaling pathways of the two blood pressure-regulating hormones angiotensin II and aldosterone are presented, focusing on both their physiological effects and the change of signaling owing to the action of increased concentrations or prolonged exposure. When present in high concentrations, both angiotensin II and aldosterone, as various other endogenous substances, activate NADPH oxidase, which produces superoxide. In this review the generation of superoxide anions and hydrogen peroxide in cells stimulated with angiotensin II or aldosterone, as well as the subsequently induced signaling processes and DNA damage is discussed.


Subject(s)
Aldosterone , Angiotensin II/physiology , Blood Pressure/physiology , Hydrogen Peroxide , Signal Transduction , Superoxides , Aldosterone/metabolism , Aldosterone/physiology , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , DNA Damage/drug effects , Endothelium, Vascular/metabolism , Humans , Hydrogen Peroxide/metabolism , Mice , NADPH Oxidases/metabolism , NADPH Oxidases/physiology , Oxidation-Reduction , Oxidative Stress , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Superoxides/metabolism
18.
Clin Lab Sci ; 23(1): 39-50, 2010.
Article in English | MEDLINE | ID: mdl-20218094

ABSTRACT

From hot dogs to Hashimoto's and inheritance to inactivity, many "entrance ramps" converge onto the "Highway to Obesity", each contributing caloric intake that exceeds expenditure. Initially, the hypothalamus regulates appetite and energy based on leptin feedback, until feedback failure increases appetite, and allows deposition of abdominal fat, metabolic dysregulation, and metabolic syndrome. Without feedback controls, progress toward obesity is unimpeded unless diet, exercise, and/or medications provide an exit ramp.


Subject(s)
Obesity/etiology , Adult , Animals , Feedback, Physiological , Genetic Predisposition to Disease , Humans , Leptin/physiology , Obesity/physiopathology , Obesity/therapy
19.
Antioxidants (Basel) ; 9(7)2020 Jul 05.
Article in English | MEDLINE | ID: mdl-32635630

ABSTRACT

In higher concentrations, the blood pressure regulating hormone angiotensin II leads to vasoconstriction, hypertension, and oxidative stress by activating NADPH oxidases which are a major enzymatic source of reactive oxygen species (ROS). With the help of knockout animals, the impact of the three predominant NADPH oxidases present in the kidney, i.e., Nox1, Nox2 and Nox4 on angiotensin II-induced oxidative damage was studied. Male wildtype (WT) C57BL/6 mice, Nox1-, Nox2- and Nox4-deficient mice were equipped with osmotic minipumps, delivering either vehicle (PBS) or angiotensin II, for 28 days. Angiotensin II increased blood pressure and urinary albumin levels significantly in all treated mouse strains. In Nox1 knockout mice these increases were significantly lower than in WT, or Nox2 knockout mice. In WT mice, angiotensin II also raised systemic oxidative stress, ROS formation and DNA lesions in the kidney. A local significantly increased ROS production was also found in Nox2 and Nox4 knockout mice but not in Nox1 knockout mice who further had significantly lower systemic oxidative stress and DNA damage than WT animals. Nox2 and Nox4 knockout mice had increased basal DNA damage, concealing possible angiotensin II-induced increases. In conclusion, in the kidney, Nox1 seemed to play a role in angiotensin II-induced DNA damage.

20.
Naunyn Schmiedebergs Arch Pharmacol ; 393(1): 121-130, 2020 01.
Article in English | MEDLINE | ID: mdl-31501914

ABSTRACT

Treatment with the chemotherapeutic agent, doxorubicin (DOX), is limited by nephrotoxicity. We investigated the possible protective effect of infliximab, a tumor necrosis factor alpha (TNF-α) inhibitor on DOX-induced nephrotoxicity. Rats were treated with a single intraperitoneal (ip) injection of DOX (17.5 mg/kg) in the absence or presence of infliximab (5 mg/kg, i.p.). Plasma and urinary markers of kidney function, oxidative stress, and inflammation were measured. Kidney and heart tissue was evaluated histopathologically. DOX-induced nephrotoxicity was confirmed by increased plasma urea, creatinine, cystatin C, neutrophil gelatinase-associated lipocalin (NGAL), and clusterin concentrations. In addition, DOX increased urinary albumin/creatinine ratio, N-acetyl-ß-D-glucosaminidase (NAG) activity, kidney injury molecule (KIM-1) concentrations, and reduced creatinine clearance. DOX significantly reduced renal antioxidants and increased plasma inflammatory markers and adiponectin concentrations. Concomitant treatment with infliximab did not significantly affect DOX-induced changes in plasma creatinine, cystatin C, or creatinine clearance. However, infliximab significantly reduced DOX-induced action on plasma urea, NGAL, clusterin, and adiponectin. Infliximab also significantly reduced urinary albumin/creatinine ratio, NAG activity, and KIM-1 concentrations, as well as the occurrence of fibrotic lesions in kidney tissue. Fibrosis detected in the heart was unchanged. In addition, infliximab reduced DOX-induced effects on plasma inflammatory markers, renal superoxide dismutase (SOD) and total antioxidant capacity. Our results show that infliximab is partially effective in mitigating DOX-induced nephrotoxicity in rats.


Subject(s)
Antineoplastic Agents , Doxorubicin , Infliximab/therapeutic use , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adiponectin/blood , Animals , Interleukin-6/blood , Kidney/drug effects , Kidney/pathology , Kidney Diseases/pathology , Male , Myocardium/pathology , Oxidative Stress/drug effects , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL