Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 286(11): 9477-88, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21199868

ABSTRACT

Sprouty-related proteins with EVH1 (enabled/vasodilator-stimulated phosphoprotein homology 1) domain (SPREDs) are inhibitors of MAPK signaling. To elucidate SPRED2 in vivo function, we characterized body homeostasis in SPRED2(-/-) mice. They showed a doubled daily water uptake, induced by elevated serum osmolality, originating from increased blood salt load. Accordingly, serum aldosterone was doubled, accompanied by augmented adrenal aldosterone synthase (AS) expression. Surprisingly, serum vasopressin (AVP) was unaltered, and, as evidenced by halved angiotensin II (Ang II) levels, the renin angiotensin system (RAS) was down-regulated. Adrenocorticotropic hormone (ACTH) was significantly elevated in SPRED2(-/-) mice, together with its secretagogue corticotropin-releasing hormone (CRH) and its downstream target corticosterone. ERK phosphorylation in brains was augmented, and hypothalamic CRH mRNA levels were elevated, both contributing to the increased CRH release. Our data were supported by CRH promoter reporter assays in hypothalamic mHypoE-44 cells, revealing a SPRED-dependent inhibition of Ets (ERK/E-twenty-six)-dependent transcription. Furthermore, SPRED suppressed CRH production in these cells. In conclusion, our study suggests that SPRED2 deficiency leads to an increased MAPK signaling, which results in an augmented CRH promoter activity. The subsequent CRH overproduction causes an up-regulation of downstream hypothalamic-pituitary-adrenal (HPA) hormone secretion. This constitutes a possible trigger for the observed compulsive grooming in SPRED2(-/-) mice and may, together with hyperplasia of aldosterone-producing cells, contribute to the hyperaldosteronism and homeostatic imbalances.


Subject(s)
Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Pituitary-Adrenal System/metabolism , Repressor Proteins/metabolism , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Phosphorylation/physiology , Repressor Proteins/genetics
2.
J Biol Chem ; 283(11): 7109-16, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18184651

ABSTRACT

The mineralocorticoid receptor (MR) is important for salt homeostasis and reno-cardiovascular pathophysiology. Signaling mechanisms include, besides classical genomic pathways, nongenomic pathways with putative pathophysiological relevance involving the mitogen-activated protein kinases ERK1/2. We determined the MR domains required for nongenomic signaling and their potential to elicit pathophysiological effects in cultured cells under defined conditions. The expression of full-length human MR or truncated MR consisting of the domains CDEF (MR CDEF), DEF (MR DEF), or EF (MR EF) renders cells responsive for the MR ligand aldosterone with respect to nongenomic ERK1/2 phosphorylation, whereas only full-length MR and MR CDEF conferred genomic responsiveness. ERK1/2 phosphorylation depends on the EGF receptor and cSRC kinase. MR EF expression is sufficient to evoke the aldosterone-induced increase of collagen III levels, similar to full-length MR expression. Our data suggest that nongenomic MR signaling is mediated by the EF domains and present the first proof of principle showing that nongenomic signaling can be sufficient for some pathophysiological effects. The minimum amino acid motif required for nongenomic MR signaling and its importance in various effects have yet to be determined.


Subject(s)
Aldosterone/metabolism , Collagen/metabolism , Gene Expression Regulation , Receptors, Mineralocorticoid/chemistry , Aldosterone/pharmacology , Amino Acid Motifs , Amino Acids/chemistry , Animals , Collagen/chemistry , Cricetinae , Humans , Ligands , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Protein Structure, Tertiary , Receptors, Mineralocorticoid/metabolism , Signal Transduction
3.
Am J Physiol Endocrinol Metab ; 292(6): E1790-800, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17311890

ABSTRACT

Aldosterone plays a key role in cardiovascular and renal injury. The underlying mechanisms are not completely understood. Because the epidermal growth factor receptor (EGFR) is involved in the development of fibrosis and vascular dysfunction, upregulation of EGFR expression by aldosterone-bound mineralocorticoid receptor (MR) is an attractive hypothesis. We investigated the effect of aldosterone on EGFR expression in the aorta of adrenalectomized rats and in human aorta smooth muscle cells (HAoSMC) in primary culture. Aldosterone, but not dexamethasone, stimulated EGFR expression in vivo in the aorta as well as in HAoSMC. EGFR degradation was not affected. Aldosterone-induced EGFR expression in HAoSMC was dose dependent and prevented by spironolactone. Furthermore, incubation of HAoSMC with aldosterone led to enhanced EGF-induced ERK1/2 phosphorylation and an EGFR-dependent increase in media fibronectin. EGFR promoter reporter gene assay as well as chromatin immunoprecipitation data indicate that MR interacts with the EGFR promoter. With deletion constructs we gained evidence that this interaction takes place between the hMR and the EGFR promoter regions 316-163 (stronger activation site, EC50 approximately 1.0 nM) and 163-1 (weaker activation site, EC50 approximately 0.7 nM), which do not comprise canonical glucocorticoid response elements and are not activated by the human glucocorticoid receptor. The interactions require in part the NH2-terminal domains of MR. ELISA-based transcription factor DNA binding assay with in vitro synthesized hMR suggest direct binding to region 163-1. Our results indicate that aldosterone leads to enhanced EGFR expression via an interaction with the EGFR promoter, which is MR specific and could contribute to the aldosterone-induced increase in fibronectin abundance.


Subject(s)
Aldosterone/physiology , Aorta/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic/physiology , Receptors, Mineralocorticoid/metabolism , Adrenalectomy , Aldosterone/pharmacology , Animals , Cells, Cultured , Chromatin , DNA/metabolism , DNA Fragmentation , Fibronectins/metabolism , Humans , Immunoprecipitation , Ligands , Male , Protein Structure, Tertiary/physiology , Rats , Rats, Wistar , Receptors, Mineralocorticoid/genetics , Tunica Media/cytology , Tunica Media/metabolism
4.
Pediatr Nephrol ; 22(6): 798-803, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17273862

ABSTRACT

Chloroacetaldehyde (CAA) is formed in the body from the chemotherapeutically used drug ifosfamide (IFO). CAA leads to cell death in proximal tubule cells mainly through the mechanism of necrosis rather than apoptosis. During chemotherapy, 2-mercaptosulfonic acid (mesna) is used with IFO to protect the urothel from cell damage. Little is known of the effect of mesna on renal proximal tubule cells, the primary site of damage after IFO treatment. Mesna contains a sulfhydryl (SH) group. To clarify whether SH-group-containing molecules can prevent CAA-induced cell death, we studied the effect of mesna and cysteine on necrosis, apoptosis, and protein content in a human proximal tubule-derived cell line (IHKE cells) treated with CAA. Both substances prevented CAA-induced necrotic cell death and protein loss and restored CAA-inhibited caspase-3 activity. CAA also prevented cisplatin-induced apoptosis. This inhibition was reversible in the presence of glutathione (GSH). We conclude that SH-containing molecules can protect proximal tubule cells from cell death because they interact with CAA before CAA can disturb other important cellular SH groups. A sufficient supply of intra- and extracellular SH groups during IFO chemotherapy may therefore have the ability to protect renal tubule cells from cell death.


Subject(s)
Acetaldehyde/analogs & derivatives , Apoptosis/drug effects , Cysteine/pharmacology , Kidney Tubules, Proximal/drug effects , Mesna/pharmacology , Protective Agents/pharmacology , Acetaldehyde/toxicity , Cell Line , Dose-Response Relationship, Drug , Drug Antagonism , Drug Therapy, Combination , Glutathione/pharmacology , Humans , Kidney Tubules, Proximal/pathology , Necrosis/chemically induced
5.
Pflugers Arch ; 454(3): 403-13, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17242955

ABSTRACT

The importance of aldosterone for cardiovascular diseases is well established. Most of the adverse effects seem to originate from its ability to produce vascular injury, including fibrosis. It is currently under debate whether aldosterone per se is able to induce fibrosis or whether it acts as a cofactor under pathological conditions. We tested whether aldosterone per se and in the presence of reactive oxygen stress (H(2)O(2)) enhances collagen abundance in human aortic smooth muscle cell (HAoSMC) media in primary culture and, if so, by which means. Collagen abundance, as well as epidermal growth factor receptor (EGFR) expression and ERK1/2 phosphorylation, was investigated by ELISA and Western blot. Collagenase activity and H(2)O(2) formation were determined by fluorometry and luminometry. Aldosterone alone did not affect collagen abundance but potentiated the stimulatory effect of low concentrations of H(2)O(2) (1-10 micromol/l). This effect disappeared when shedding of membrane-bound EGFR ligands was prevented by GM6001. EGFR expression and cellular EGF responsiveness were enhanced by aldosterone. Inhibition of the EGFR kinase (tyrphostin AG1478) prevented the increase of collagen. The increase in collagen abundance was prevented by blockade of the mineralocorticoid receptor (MR) and could be reproduced by MR transfection into Chinese hamster ovary cells. We conclude that aldosterone sensitizes HAoSMC for H(2)O(2)-induced increase of collagen abundance at least in part by enhanced EGFR expression.


Subject(s)
Aldosterone/pharmacology , Collagen/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Base Sequence , Cells, Cultured , Collagenases/metabolism , ErbB Receptors/metabolism , Gelatinases/metabolism , Homeostasis , Humans , Hydrogen Peroxide/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Receptors, Mineralocorticoid/genetics
6.
Mol Cell Endocrinol ; 264(1-2): 35-43, 2007 Jan 29.
Article in English | MEDLINE | ID: mdl-17113706

ABSTRACT

The mineralocorticcoid receptor (MR) plays an important role in salt and water homeostasis as well as during cardiovascular and renal fibrosis but little is known regarding its modulation by other signaling pathways. To investigate a possible modulation under controlled conditions we used human embryonic kidney (HEK) cells (devoid of endogenous MR) transfected with the human MR and measured transactivation with a GRE-SEAP-reporter construct. MR was compared to the glucocorticoid receptor (GR) as well as to MR lacking the N-terminal domains AB (MR(CDEF)). Chelation of cytosolic Ca2+ enhanced MR activity and SGK1-expression, whereas elevation of cytosolic Ca2+ with ionomycin or thapsigargin reduced MR activity. GR activity was not affected by ionomycin or thapsigargin. MR(CDEF) activity was not affected by chelation or elevation of cytosolic Ca2+. Inhibition of ERK1/2 activation by U0126 or activation of PKA by cAMP, previously shown to modulate MR and GR activity, did not affect MR(CDEF) activity either. H2O2<500micromol/l did not affect basal nor hormone-induced reporter activity. Higher concentrations exerted the same relative inhibitory effect on GRE-SEAP-activity under basal conditions as in the presence of aldosterone-stimulated MR and elicited cytotoxic effects. Our data indicate that the genomic function of MR can be modulated by cytosolic Ca2+, PKA and ERK1/2 via an interaction with the AB-domain. H2O2 seems not to affect relative MR activity directly under our experimental conditions.


Subject(s)
Calcium Signaling/drug effects , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , Receptors, Mineralocorticoid/biosynthesis , Response Elements/physiology , Calcium/metabolism , Calcium Signaling/physiology , Cell Line , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , Kidney/cytology , Kidney/metabolism , Protein Kinases/metabolism , Receptors, Glucocorticoid/biosynthesis
7.
Kidney Blood Press Res ; 29(5): 280-93, 2006.
Article in English | MEDLINE | ID: mdl-17035713

ABSTRACT

Chloroacetaldehyde (CAA) is a metabolite of the alkylating agent ifosfamide (IFO) and putatively responsible for renal damage following anti-tumor therapy with IFO. Depletion of sulfhydryl (SH) groups has been reported from cell culture, animal and clinical studies. In this work the effect of CAA on human proximal tubule cells in primary culture (hRPTEC) was investigated. Toxicity of CAA was determined by protein content, cell number, LDH release, trypan blue exclusion assay and caspase-3 activity. Free thiols were measured by the method of Ellman. CAA reduced hRPTEC cell number and protein, induced a loss in free intracellular thiols and an increase in necrosis markers. CAA but not acrolein inhibited the cysteine proteases caspase-3, caspase-8 and cathepsin B. Caspase activation by cisplatin was inhibited by CAA. In cells stained with fluorescent dyes targeting lysosomes, CAA induced an increase in lysosomal size and lysosomal leakage. The effects of CAA on cysteine protease activities and thiols could be reproduced in cell lysate. Acidification, which slowed the reaction of CAA with thiol donors, could also attenuate effects of CAA on necrosis markers, thiol depletion and cysteine protease inhibition in living cells. Thus, CAA directly reacts with cellular protein and non-protein thiols, mediating its toxicity on hRPTEC. This effect can be reduced by acidification. Therefore, urinary acidification could be an option to prevent IFO nephropathy in patients.


Subject(s)
Acetaldehyde/analogs & derivatives , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/toxicity , Ifosfamide/metabolism , Ifosfamide/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Sulfhydryl Compounds/toxicity , Sulfhydryl Reagents/metabolism , Acetaldehyde/chemistry , Acetaldehyde/metabolism , Biomarkers , Caspase 3/metabolism , Caspase 8/metabolism , Cathepsin B/metabolism , Cell Count , Cell Death , Cells, Cultured , Coloring Agents , Cysteine Endopeptidases/metabolism , Humans , Kidney Diseases/enzymology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , L-Lactate Dehydrogenase/metabolism , Microscopy, Fluorescence , Necrosis , Sulfhydryl Compounds/chemistry , Sulfhydryl Reagents/chemistry , Trypan Blue , gamma-Glutamyltransferase/metabolism
8.
Pediatr Nephrol ; 21(1): 60-7, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16267686

ABSTRACT

Ifosfamide (ifo) is a commonly used drug in chemotherapy. It is metabolized to acrolein (acro) and chloroacetaldehyde (CAA), which are thought to be responsible for renal side effects. We studied the effects of ifo and cyclophosphamide (cyclo) as well as their metabolites, acro and CAA, on cellular protein content, necrosis, apoptosis and cytosolic calcium concentration using a human proximal tubule cell line. The protein content decreased during acro or CAA administration (15 to 300 micromol/l), but not during ifo or cyclo exposure over a time period of up to 72 h. Mild apoptosis was induced only by high acro (150, 300 micromol/l) and low CAA concentrations (15, 75 micromol/l) and only in a narrow time window (24 h). Necrosis was increased after exposure to acro or CAA at all concentrations. CAA was more potent than acro. Ifo and cyclo did not induce necrosis or apoptosis. Glutathione abolished CAA-induced cell death. Cytosolic calcium concentrations increased after acro or CAA administration and showed an oscillating pattern. Cytosolic Ca(2+) chelation did not prevent necrosis. We conclude that neither ifo nor cyclo induce cell damage, but that their metabolites acro and CAA induce cell death. This cell death occurs mainly by necrosis and not by apoptosis.


Subject(s)
Acetaldehyde/analogs & derivatives , Acrolein/pharmacology , Glutathione/pharmacology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Acetaldehyde/pharmacology , Cell Death/drug effects , Cells, Cultured , Humans , Oxidation-Reduction
9.
Kidney Int ; 68(5): 2029-41, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16221203

ABSTRACT

BACKGROUND: Renal damage following chemotherapy with ifosfamide is attributed to the metabolic activation of the drug and the generation of chloroacetaldehyde (CAA). Little is known about the mechanism by which CAA impairs renal function. In this study the effect of CAA on intracellular Ca(2+) homeostasis in human renal proximal tubule cells (RPTEC) in primary culture was investigated. METHODS: Intracellular Ca(2+) was measured using the Ca(2+)-sensitive dye fura-2. Cell viability was determined by protein content and cell number. Oncotic and apoptotic cell death was assayed using trypan blue exclusion, caspase-3 activity, and 4',6-diamino-2-phenylindole (DAPI) staining. RESULTS: CAA (1.5 to 150 micromol/L) induced sustained elevations of intracellular free calcium ([Ca(2+)](i)) from 75 +/- 3 nmol/L to maximal 151 +/- 6 nmol/L. This effect was dependent on extracellular Ca(2+), but not Ca(2+) entry. The rise in [Ca(2+)](i) mediated by CAA could be attributed to inhibition of Na(+)-dependent extrusion of intracellular Ca(2+), indicating an inhibitory action of CAA on Na(+)/Ca(2+) exchange. Modulation of protein kinase A (PKA), but not protein kinase C (PKC) blunted the effect of CAA. Thus, CAA seems to inhibit Na(+)/Ca(2+) exchange by interaction with cyclic adenosine monophosphate (cAMP)-PKA-signaling. A 48-hour exposure to 15 micromol/L CAA significantly reduced cell number and protein content of RPTEC by induction of necrosis. This effect of 15 micromol/L CAA could be overcome by coadministration of the intracellular Ca(2+) chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester (BAPTA-AM). CONCLUSION: First, CAA inhibits the Na+/Ca2+-exchanger. Second, this effect is dependent on PKA. Third, CAA induces necrotic rather than apoptotic cell death. Finally, disturbed Ca(2+) homeostasis via Na(+)/Ca(2+) exchange contributes to the nephrotoxic action of CAA in RPTEC.


Subject(s)
Acetaldehyde/analogs & derivatives , Antineoplastic Agents, Alkylating/toxicity , Calcium Signaling/drug effects , Ifosfamide/toxicity , Kidney Diseases/chemically induced , Kidney Tubules, Proximal/drug effects , Acetaldehyde/metabolism , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Cation Transport Proteins/metabolism , Cell Line , Cytosol/drug effects , Cytosol/metabolism , Extracellular Space/metabolism , Homeostasis/drug effects , Humans , Hydrogen-Ion Concentration , Kidney Diseases/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Plasma Membrane Calcium-Transporting ATPases , RNA, Messenger/analysis , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium/metabolism , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism
10.
Mol Endocrinol ; 19(7): 1697-710, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15761031

ABSTRACT

The steroid hormone aldosterone is important for salt and water homeostasis as well as for pathological tissue modifications in the cardiovascular system and the kidney. The mechanisms of action include a classical genomic pathway, but physiological relevant nongenotropic effects have also been described. Unlike for estrogens or progesterone, the mechanisms for these nongenotropic effects are not well understood, although pharmacological studies suggest a role for the mineralocorticoid receptor (MR). Here we investigated whether the MR contributes to nongenotropic effects. After transfection with human MR, aldosterone induced a rapid and dose-dependent phosphorylation of ERK1/2 and c-Jun NH2-terminal kinase (JNK) 1/2 kinases in Chinese hamster ovary or human embryonic kidney cells, which was reduced by the MR-antagonist spironolactone and involved cSrc kinase as well as the epidermal growth factor receptor. In primary human aortic endothelial cells, similar results were obtained for ERK1/2 and JNK1/2. Inhibition of MAPK kinase (MEK) kinase but not of protein kinase C prevented the rapid action of aldosterone and also reduced aldosterone-induced transactivation, most probably due to impaired nuclear-cytoplasmic shuttling of MR. Cytosolic Ca2+ was increased by aldosterone in mock- and in human MR-transfected cells to the same extend due to Ca2+ influx, whereas dexamethasone had virtually no effect. Spironolactone did not prevent the Ca2+ response. We conclude that some nongenotropic effects of aldosterone are MR dependent and others are MR independent (e.g. Ca2+), indicating a higher degree of complexity of rapid aldosterone signaling. According to this model, we have to distinguish three aldosterone signaling pathways: 1) genomic via MR, 2) nongenotropic via MR, and 3) nongenotropic MR independent.


Subject(s)
Aldosterone/physiology , Mitogen-Activated Protein Kinases/metabolism , Receptors, Mineralocorticoid/metabolism , Signal Transduction , Aldosterone/pharmacology , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Cytosol/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Homeostasis , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Receptors, Mineralocorticoid/genetics
11.
Toxicol Sci ; 85(1): 735-42, 2005 May.
Article in English | MEDLINE | ID: mdl-15716484

ABSTRACT

Cisplatin is a widely used chemotherapeutic agent. Here we show that cisplatin induces apoptosis in renal collecting duct-derived cells (MDCK-C7 cells, resembling principal cells) in a dose-dependent manner. Additionally, we studied the role of mitochondria in this process by inhibition of the mitochondrial respiratory chain, the F1F(o)-ATP synthase or by uncoupling. The role of intra- and extracellular pH in apoptosis induction was investigated. Activation of caspase-3 and DNA ladder formation were used to monitor the apoptotic response. When cells were incubated with inhibitors of the mitochondrial respiratory chain or an inhibitor of the ATP-synthase, cisplatin-induced apoptosis was markedly enhanced. Mitochondrial blockade led to enhanced production of lactic acid. Also, anoxia potentiated the cisplatin-induced caspase-3 activation. Neither intra- nor extracellular pH had an influence on caspase-3 activation at low cisplatin concentrations. Acidic conditions (pH 6.8) potentiated the caspase-3 activation when high (100 microM) cisplatin concentrations were used. We demonstrate that intact mitochondria are important to prevent cisplatin-induced apoptosis in MDCK-C7 cells and that acidic conditions can aggravate the toxic effects of cisplatin.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cisplatin/toxicity , Kidney Tubules, Collecting/drug effects , Mitochondria/drug effects , Animals , Apoptosis/physiology , Caspase 3 , Caspases/metabolism , Cell Hypoxia , Cell Line , Cell Survival/drug effects , Dogs , Hydrogen-Ion Concentration , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Mitochondria/enzymology
12.
Toxicol In Vitro ; 19(1): 135-43, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15582364

ABSTRACT

The body may be exposed simultaneously to more than one nephrotoxic substance and to measure the effects of the great number of possible combinations of nephrotoxins will rapidly become a great challenge when using the traditional methods. Therefore, we developed a rapid and cost-efficient method to screen the apoptotic potential of combinations of known cell- or nephrotoxic substances as ochratoxin A (OTA), cisplatin, cadmium, H(2)O(2), and amphotericin B on renal epithelial cell lines. The cells were seeded in 96-well plates and the apoptotic and necrotic potential of different combinations of nephrotoxins was determined. We found different results for the combinations used: depending on the concentrations of the various substances, antagonistic, additive, or potentiating effects on caspase-3 activity were found after co-exposure to OTA. We conclude that the co-exposure of renal cells to OTA with other substances can enhance or reduce the apoptotic potential of one substance alone depending on the substance, the concentration and on the cell line investigated. A "harmless" substance can thus convert to a potent cell toxic substance when combined with OTA or vice versa. The underlying mechanisms of the synergistic effects remain unknown.


Subject(s)
Apoptosis/drug effects , Kidney Tubules, Proximal/drug effects , Mycotoxins/toxicity , Ochratoxins/toxicity , Toxicity Tests/methods , Cadmium/toxicity , Caspase 3 , Caspases/metabolism , Cell Line, Transformed , Cisplatin/toxicity , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Humans , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/pathology , Proteins/metabolism , Toxicity Tests/economics , Trypan Blue/metabolism
13.
Am J Physiol Renal Physiol ; 287(3): F469-73, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15113744

ABSTRACT

Proximal tubular receptor-mediated endocytosis (RME) of filtered proteins prevents proteinuria. Pharmacological and genetic studies in cultured opossum kidney cells have shown that the apical Na(+)/H(+) exchanger isoform 3 (NHE3) supports RME by interference with endosomal pH homeostasis and endocytic fusion events. However, it is not known whether NHE3 also supports proximal tubular RME in vivo. We analyzed proximal tubular protein reabsorption by microinfusion experiments in rats and investigated renal protein excretion in NHE3 knockout (Nhe3 -/-) mice. Inhibition of NHE3 by EIPA or S-3226 reduced the fractional reabsorption of [(14)C]cytochrome c by approximately 50% during early proximal microinfusion. During early distal microinfusion, no protein reabsorption could be detected. Urinary protein excretion of Nhe3 -/- or heterozygous mutant mice was significantly higher compared with wild-type mice. SDS-PAGE analysis of urinary proteins revealed that Nhe3 -/- animals excreted proteins the size of albumin or smaller. Thus a reduction in NHE3 activity or abundance causes tubular proteinuria. These data show that NHE3 supports proximal tubular RME of filtered proteins in vivo.


Subject(s)
Kidney Tubules, Proximal/metabolism , Sodium-Hydrogen Exchangers/metabolism , Absorption/physiology , Animals , Homeostasis/physiology , Hydrogen-Ion Concentration , Male , Mice , Mice, Knockout , Proteins/metabolism , Proteinuria/metabolism , Rats , Rats, Wistar , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics
14.
Cell Physiol Biochem ; 14(1-2): 47-56, 2004.
Article in English | MEDLINE | ID: mdl-14976406

ABSTRACT

Ochratoxin A (OTA) is a potent nephrotoxin and suspected to be involved in the etiology of Balkan endemic nephropathy. Nanomolar concentrations of this mycotoxin induce apoptosis in renal collecting duct-derived cells (MDCK-C7 cells, resembling principal cells). We studied the role of mitochondria in this process by inhibition of the mitochondrial respiratory chain, the F1FO-ATP synthase or by uncoupling. Also, the role of intra- and extracellular pH in apoptosis induction was investigated. Activation of caspase-3 and DNA ladder formation were used to monitor the apoptotic response. When cells were incubated with inhibitors of the mitochondrial respiratory chain or an inhibitor of the ATP-synthase, OTA-induced apoptosis was enhanced dramatically. Also, mitochondrial uncoupling potentiated the effects of OTA. OTA-induced apoptosis was not dependent on a decrease of the mitochondrial potential. Mitochondrial blockade led to medium acidification due to enhanced production of lactic acid. Artificial extracellular acidification potentiated OTA-induced caspase-3 activation. Artificial extracellular alkalization had no influence on caspase-3 activity. Intracellular pH after 24 hours exposure to inhibitors of mitochondria or acidic or alkaline media did not correlate with caspase-3 activity but correlated with caspase-3 activity when OTA was present: acidic intracellular pH (pHin) was associated with higher caspase-3 activity as compared to alkaline pHin. We conclude that extra- and intracellular pH are important factors in OTA-induced apoptosis in MDCK-C7 cells. The physiologically changing pH conditions in the collecting duct can thus alter or even aggravate the toxic effects of OTA.


Subject(s)
Apoptosis/physiology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/metabolism , Mitochondria/physiology , Ochratoxins/toxicity , Acids/pharmacology , Alkalies/pharmacology , Animals , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Line , Clone Cells/drug effects , Clone Cells/metabolism , DNA/metabolism , Dogs , Hydrogen-Ion Concentration/drug effects , Kidney Tubules, Collecting/cytology , Lactic Acid/metabolism , Mitochondria/drug effects , Uncoupling Agents/pharmacology
15.
Am J Physiol Renal Physiol ; 286(6): F1226-31, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14749256

ABSTRACT

Aldosterone enhances Na(+) reabsorption via epithelial Na(+) channels (ENaC). Aldosterone also stimulates the protein kinase ERK1/2- and the epidermal growth factor (EGF) receptor (EGFR)-signaling pathway. Yet EGF and ERK1/2 are known inhibitors of ENaC-mediated Na(+) reabsorption. In the present study, using the well-established Madin-Darby canine kidney C7 cell line, we tested the hypothesis that EGFR represents a negative-feedback control for chronic aldosterone-induced Na(+) reabsorption [amiloride-inhibitable short-circuit current (I(sc))]. Mineralocorticoid receptor expression was confirmed by RT-PCR and Western blot analysis. Aldosterone enhanced ERK1/2 phosphorylation in an EGFR-dependent way. Furthermore, aldosterone stimulated EGFR expression. Aldosterone (10 nmol/l) induced a small transient increase in I(sc) under control conditions. Inhibition of ERK1/2 phosphorylation with U-0126 (10 micromol/l) stimulated I(sc), indicating constitutive ENaC inhibition. Aldosterone exerted a significantly larger effect in the presence of U-0126 than without U-0126. EGF (10 microg/l) inhibited I(sc), whereas inhibition of EGFR kinase by tyrphostin AG-1478 (100 nmol/l) enhanced I(sc). Aldosterone was more effective in the presence of AG-1478 than without AG-1478. In summary, we propose that the EGFR-signaling cascade can serve as a negative-feedback control to limit the effect of aldosterone-induced Na(+) reabsorption.


Subject(s)
Aldosterone/pharmacology , ErbB Receptors/physiology , Sodium/metabolism , Animals , Blotting, Western , Cells, Cultured , Dogs , Enzyme-Linked Immunosorbent Assay , Epithelial Sodium Channels , Feedback/physiology , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sodium Channels/metabolism , Sodium Channels/physiology
16.
J Physiol ; 552(Pt 2): 471-81, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14561830

ABSTRACT

Transforming growth factor (TGF)-beta1 is a member of a superfamily of multifunctional cytokines involved in several pathological processes of the kidney, including fibrogenesis, apoptosis and epithelial-mesenchymal transition. These events lead to tubulointerstitial fibrosis and glomerulosclerosis. Less is known about TGF-beta1-induced alterations of cell function. An important function of proximal tubular cells is reabsorption of filtered proteins, including albumin, via megalin-cubilin-dependent receptor-mediated endocytosis. In this study we used a well established cell culture model (proximal-tubule-derived opossum kidney (OK) cells) in order to test the hypothesis that TGF-beta1 reduces megalin-cubilin-mediated endocytosis. Previously we have shown that albumin endocytosis in OK cells is mediated by megalin/cubulin. TGF-beta1 led to a time- and dose-dependent downregulation of megalin-cubilin-mediated endocytosis without affecting two other transport systems tested. Binding, internalization and intracellular trafficking of the ligand albumin were affected. Decreased binding resulted from reduced cubilin and megalin expression in the 200 000 g membrane fraction. The underlying mechanism of TGF-beta1 action does not involve mitogen-activated protein kinases, protein kinase C or A, or reactive oxygen species. In contrast, TGF-beta1-induced downregulation of megalin-cubilin-mediated endocytosis was sensitive to inhibition of translation and transcription and was preceded by Smad2 and 3 phosphorylation. Dominant negative Smad2/3 constructs prevented the effect of TGF-beta1. In conclusion our data indicate that enhanced levels of TGF-beta1 occurring in various nephropathies can lead to downregulation of megalin-cubilin-dependent endocytosis. Probably, TGF-beta1 leads to Smad2- and Smad3-dependent expression of negative regulators of receptor-mediated endocytosis.


Subject(s)
Albumins/metabolism , Endocytosis/physiology , Kidney Tubules, Proximal/physiology , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Opossums/physiology , Receptors, Cell Surface/physiology , Transforming Growth Factor beta/physiology , Animals , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , Collagen/metabolism , DNA-Binding Proteins/metabolism , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Epithelium/physiology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Smad2 Protein , Smad3 Protein , Trans-Activators/metabolism
17.
J Biol Chem ; 278(44): 43060-6, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12939263

ABSTRACT

The steroid hormone aldosterone plays an important role during pathological tissue modifications, similar to cardiovascular or renal fibrosis. The underlying mechanisms for the pathological actions are not understood. Interaction of aldosterone with the epidermal growth factor (EGF) receptor is an attractive hypothesis to explain pathological tissue remodeling elicited by aldosterone, because (i) mineralocorticoids can sensitize cells for EGF, (ii) mineralocorticoid receptor (MR)-antagonists reduce EGFR-mRNA expression, (iii) EGFR itself supports the development of cardiovascular or renal fibrosis, and (iv) signaling elements involved in the pathological action of aldosterone (similar to ERK1/2 or NFkB) are typical downstream modules during EGF signaling. In addition, an interaction of aldosterone and EGF with respect to ERK1/2 activation has been described. Here we show that aldosterone stimulates EGFR expression in renal tissue of adrenalectomized rats and in human renal primary cell cultures. Furthermore, Chinese hamster ovary (CHO) cells normally devoid of EGFR or MR express EGFR after transfection with human MR (CHO-MR cells) but not after transfection with human glucocorticoid receptor (CHO-GR cells). In CHO-MR cells, EGFR-expression is up-regulated by aldosterone and inhibited by spironolactone. CHO-MR cells but not CHO-GR cells respond with ERK1/2 phosphorylation to EGF exposure. The responsiveness to other peptide hormones was virtually not affected. These data suggest that EGFR is an aldosterone-induced protein and is involved in the manifold (patho)biological actions of aldosterone.


Subject(s)
Aldosterone/physiology , ErbB Receptors/biosynthesis , Aldosterone/metabolism , Animals , Base Sequence , Blotting, Western , CHO Cells , Cells, Cultured , Cloning, Molecular , Cricetinae , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Humans , Kidney/cytology , Kidney/metabolism , Male , Microscopy, Fluorescence , Mineralocorticoids/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phosphorylation , Promoter Regions, Genetic , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Time Factors , Transfection , Up-Regulation
18.
Kidney Int Suppl ; (84): S103-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12694321

ABSTRACT

BACKGROUND: Interstitial fibrosis is of major importance for the deterioration of renal function, leading to uremia. Interaction of filtered proteins with proximal tubular cells is important for the onset and development of tubulointerstitial damage. METHODS: We investigated the effects of protein endocytosis on collagen homeostasis and signaling pathways of proximal tubule-derived cells (OK cells, LLC-PK1 cells), which express the endocytic machinery typical for the proximal tubule (megalin and cubilin), and compared it to renal epithelial cells with low endocytic activity (MDCK, IHKE1, NHE3-deficient OK cells). Collagen homeostasis was assessed by proline incorporation, ELISA, and Western blot. Matrix metalloproteinase (MMP) activity was assessed by gelatinase assay. Signaling pathways were monitored by reporter gene assay. RESULTS: Albumin, glycated albumin, fatty acid-free albumin, or globulins led to an increase of secreted collagen (types I, III, and IV) in OK and LLC-PK1 cells. In cells with low protein uptake activity, albumin exposure inhibited collagen secretion. Western blot analysis showed an increase of cellular collagen. MMP activity was significantly decreased by albumin exposure. Furthermore, albumin exposure led to activation of the NF-kappa B-, AP1-, NFAT-, SRE-, and CRE-pathways. Inhibition of NF-kappa B, PKC, or PKA partially reversed the effects of albumin. In addition, inhibition of albumin endocytosis reduced collagen secretion and activation of the signaling pathways. Discussion. The data show that endocytic uptake of proteins disturbs collagen homeostasis in proximal tubular cells. This disturbed matrix homeostasis probably supports the progression of interstitial fibrosis, which is of importance for the development of renal insufficiency.


Subject(s)
Albumins/pharmacokinetics , Collagen/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Animals , Blotting, Western , Collagen/analysis , Collagenases , Enzyme-Linked Immunosorbent Assay , Gelatinases/metabolism , Genes, Reporter , Homeostasis/physiology , LLC-PK1 Cells , Opossums , Proline/pharmacokinetics , Signal Transduction/physiology , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Swine
19.
Kidney Int ; 63(5): 1725-35, 2003 May.
Article in English | MEDLINE | ID: mdl-12675848

ABSTRACT

BACKGROUND: Nephrotoxic substances like cisplatin or ochratoxin A (OTA) induce cell death in human proximal tubule-derived cells (IHKE cells). Mitochondria play a significant role in apoptosis and loss of their function may influence OTA- or cisplatin-induced apoptosis. Extracellular pH also plays an important role in tumor genesis. Therefore, we investigated the role of mitochondria and intra- and extracellular pH on cell death induction by cisplatin or OTA. METHODS: IHKE cells were incubated in the presence of OTA or cisplatin, together with inhibitors of the mitochondrial metabolism, and the activity of caspase-3 was measured and DNA laddering was monitored. Adenosine triphosphate (ATP) content of the cells, lactate release into the media, and glucose consumption was determined. In addition, media and cells were acidified or alkalized artificially to investigate the effect of intra- and extracellular pH on cell death induction. Cytochrome C was immunodetected in cellular compartments. RESULTS: Inhibition of the mitochondrial function reduced OTA- or cisplatin-induced cell death and led to considerable lactic acid production and extracellular acidification. Intra- and extracellular acidification prevented cells from cell death induced by OTA or cisplatin. No cytochrome C release from mitochondria could be detected during 24 hours of exposure to OTA or cisplatin. CONCLUSION: We conclude that OTA- or cisplatin-induced cell death is dependent on functional and intact, ATP-producing mitochondria and that intra- and extracellular pH is crucial for induction of cell death in IHKE cells.


Subject(s)
Acidosis/pathology , Apoptosis/physiology , Epithelial Cells/cytology , Kidney Tubules, Proximal/cytology , Mitochondria/physiology , Acidosis/metabolism , Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Buffers , Carcinogens/pharmacology , Caspase 3 , Caspases/metabolism , Cell Line, Transformed , Cisplatin/pharmacology , Cytochromes c/metabolism , DNA/analysis , Electron Transport/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Space/metabolism , Glucose/metabolism , Humans , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Mitochondria/drug effects , Mitochondrial ADP, ATP Translocases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Ochratoxins/pharmacology , Uncoupling Agents/pharmacology
20.
J Biol Chem ; 277(48): 45892-7, 2002 Nov 29.
Article in English | MEDLINE | ID: mdl-12244120

ABSTRACT

The epidermal growth factor (EGF) regulates cell proliferation, differentiation, and ion transport using ERK1/2 as a downstream effector. Furthermore, the EGF receptor (EGFR) is involved in signaling by G-protein-coupled receptors, growth hormone, and cytokines via transactivation. It has been suggested that steroids interact with peptide hormones. Previously, we have shown that aldosterone modulates EGF responses in Madin-Darby canine kidney cells (Gekle, M., Freudinger, R., Mildenberger, S., and Silbernagl, S. (2002) Am. J. Physiol. 282, F669-F679). Here, we tested the hypothesis that human EGFR-1 can confer alternative aldosterone responsiveness with respect to ERK1/2 phosphorylation to Chinese hamster ovary cells, which do not express EGFR. Wild-type Chinese hamster ovary cells did not respond to EGF or aldosterone. After transfection of human EGFR-1, the cells responded to EGF, but not to aldosterone. However, when submaximal concentrations of EGF were used, nanomolar concentrations of aldosterone potentiated the action of EGF within minutes, resulting in a leftward shift of the EGF dose-response curve. This was not the case in mock-transfected cells. The EGFR kinase inhibitor tyrphostin AG1478 or the MEK1/2 inhibitor U0126 completely prevented the effect. Furthermore, aldosterone enhanced Tyr phosphorylation of c-Src and EGFR, and an inhibitor of cytosolic tyrosine kinases (4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyriociaine) prevented the action of aldosterone. Our data show that aldosterone uses the EGF-EGFR-MEK1/2-ERK1/2 signaling cascade to elicit its alternative effects. In the presence of EGF, aldosterone leads to EGFR transactivation via cytosolic tyrosine kinases of the Src family.


Subject(s)
Aldosterone/metabolism , ErbB Receptors/physiology , Signal Transduction/physiology , Animals , CHO Cells , Cricetinae , Dogs , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/genetics , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...