Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Apoptosis ; 20(9): 1200-10, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26067145

ABSTRACT

In rodents, ubiquitous α1-Na(+), K(+)-ATPase is inhibited by ouabain and other cardiotonic steroids (CTS) at ~10(3)-fold higher concentrations than those effective in other mammals. To examine the specific roles of the CTS-sensitive α1S- and CTS-resistant α1R-Na(+), K(+)-ATPase isoforms, we compared the effects of ouabain on intracellular Na(+) and K(+) content, cell survival, and mitogen-activated protein kinases (MAPK) in human and rat vascular smooth muscle cells (HASMC and RASMC), human and rat endothelial cells (HUVEC and RAEC), and human and rat brain astrocytes. 6-h exposure of HASMC and HUVEC to 3 µM ouabain dramatically increased the intracellular [Na(+)]/[K(+)] ratio to the same extend as in RASMC and RAEC treated with 3000 µM ouabain. In 24, 3 µM ouabain triggered the death of all types of human cells used in this study. Unlike human cells, we did not detect any effect of 3000-5000 µM ouabain on the survival of rat cells, or smooth muscle cells from mouse aorta (MASMC). Unlike in the wild-type α1(R/R) mouse, ouabain triggered death of MASMC from α1(S/S) mouse expressing human α1-Na(+), K(+)-ATPase. Furthermore, transfection of HUVEC with rat α1R-Na(+), K(+)-ATPase protected them from the ouabain-induced death. In HUVEC, ouabain led to phosphorylation of p38 MAPK, whereas in RAEC it stimulated phosphorylation of ERK1/2. Overall, our results, demonstrate that the drastic differences in cytotoxic action of ouabain on human and rodent cells are caused by unique features of α1S/α1R-Na(+), K(+)-ATPase, rather than by any downstream CTS-sensitive/resistant components of the cell death machinery.


Subject(s)
Cardiotonic Agents/toxicity , Cell Death/drug effects , Ouabain/toxicity , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Biomarkers/metabolism , Brain/cytology , Cell Line , Epithelial Cells/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle, Smooth/cytology , Potassium/metabolism , Protein Structure, Tertiary , Rats, Sprague-Dawley , Sodium/metabolism
2.
PLoS One ; 9(11): e110597, 2014.
Article in English | MEDLINE | ID: mdl-25375852

ABSTRACT

This study examines the relative impact of canonical hypoxia-inducible factor-1alpha- (HIF-1α and Na+i/K+i-mediated signaling on transcriptomic changes evoked by hypoxia and glucose deprivation. Incubation of RASMC in ischemic conditions resulted in ∼3-fold elevation of [Na+]i and 2-fold reduction of [K+]i. Using global gene expression profiling we found that Na+,K+-ATPase inhibition by ouabain or K+-free medium in rat aortic vascular smooth muscle cells (RASMC) led to the differential expression of dozens of genes whose altered expression was previously detected in cells subjected to hypoxia and ischemia/reperfusion. For further investigations, we selected Cyp1a1, Fos, Atf3, Klf10, Ptgs2, Nr4a1, Per2 and Hes1, i.e. genes possessing the highest increments of expression under sustained Na+,K+-ATPase inhibition and whose implication in the pathogenesis of hypoxia was proved in previous studies. In ouabain-treated RASMC, low-Na+, high-K+ medium abolished amplification of the [Na+]i/[K+]i ratio as well as the increased expression of all tested genes. In cells subjected to hypoxia and glucose deprivation, dissipation of the transmembrane gradient of Na+ and K+ completely eliminated increment of Fos, Atf3, Ptgs2 and Per2 mRNAs and sharply diminished augmentation expression of Klf10, Edn1, Nr4a1 and Hes1. In contrast to low-Na+, high-K+ medium, RASMC transfection with Hif-1a siRNA attenuated increments of Vegfa, Edn1, Klf10 and Nr4a1 mRNAs triggered by hypoxia but did not impact Fos, Atf3, Ptgs2 and Per2 expression. Thus, our investigation demonstrates, for the first time, that Na+i/K+i-mediated, Hif-1α- -independent excitation-transcription coupling contributes to transcriptomic changes evoked in RASMC by hypoxia and glucose deprivation.


Subject(s)
Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia/genetics , Transcriptome , Animals , Enzyme Inhibitors/pharmacology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Ouabain/pharmacology , Rats , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism
3.
PLoS One ; 7(5): e38032, 2012.
Article in English | MEDLINE | ID: mdl-22666440

ABSTRACT

Stimulus-dependent elevation of intracellular Ca(2+) ([Ca(2+)](i)) affects the expression of numerous genes--a phenomenon known as excitation-transcription coupling. Recently, we found that increases in [Na(+)](i) trigger c-Fos expression via a novel Ca(2+) (i)-independent pathway. In the present study, we identified ubiquitous and tissue-specific [Na(+)](i)/[K(+)](i)-sensitive transcriptomes by comparative analysis of differentially expressed genes in vascular smooth muscle cells from rat aorta (RVSMC), the human adenocarcinoma cell line HeLa, and human umbilical vein endothelial cells (HUVEC). To augment [Na(+)](i) and reduce [K(+)](i), cells were treated for 3 hrs with the Na(+),K(+)-ATPase inhibitor ouabain or placed for the same time in the K(+)-free medium. Employing Affymetrix-based technology, we detected changes in expression levels of 684, 737 and 1839 transcripts in HeLa, HUVEC and RVSMC, respectively, that were highly correlated between two treatments (p<0.0001; R(2)>0.62). Among these Na(+) (i)/K(+) (i)-sensitive genes, 80 transcripts were common for all three types of cells. To establish if changes in gene expression are dependent on increases in [Ca(2+)](i), we performed identical experiments in Ca(2+)-free media supplemented with extracellular and intracellular Ca(2+) chelators. Surprisingly, this procedure elevated rather than decreased the number of ubiquitous and cell-type specific Na(+) (i)/K(+) (i)-sensitive genes. Among the ubiquitous Na(+) (i)/K(+) (i)-sensitive genes whose expression was regulated independently of the presence of Ca(2+) chelators by more than 3-fold, we discovered several transcription factors (Fos, Jun, Hes1, Nfkbia), interleukin-6, protein phosphatase 1 regulatory subunit, dual specificity phosphatase (Dusp8), prostaglandin-endoperoxide synthase 2, cyclin L1, whereas expression of metallopeptidase Adamts1, adrenomedulin, Dups1, Dusp10 and Dusp16 was detected exclusively in Ca(2+)-depleted cells. Overall, our findings indicate that Ca(2+) (i)-independent mechanisms of excitation-transcription coupling are involved in transcriptomic alterations triggered by elevation of the [Na(+)](i)/[K(+)](i) ratio. There results likely have profound implications for normal and pathological regulation of mammalian cells, including sustained excitation of neuronal cells, intensive exercise and ischemia-triggered disorders.


Subject(s)
Calcium/metabolism , Potassium/metabolism , Sodium/metabolism , Transcription, Genetic , Transcriptome , Animals , Cell Survival/drug effects , Chelating Agents/pharmacology , Enzyme Inhibitors/pharmacology , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Ouabain/pharmacology , Rats , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Transcription, Genetic/drug effects , Transcriptome/drug effects
4.
Can J Physiol Pharmacol ; 90(2): 209-17, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22300272

ABSTRACT

In the present work, we compared the outcome of hyperosmotic and isosmotic shrinkage on ion transport and protein phosphorylation in C11-MDCK cells resembling intercalated cells from collecting ducts and in vascular smooth muscle cells (VSMC) from the rat aorta. Hyperosmotic shrinkage was triggered by cell exposure to hypertonic medium, whereas isosmotic shrinkage was evoked by cell transfer from an hypoosmotic to an isosmotic environment. Despite a similar cell volume decrease of 40%-50%, the consequences of hyperosmotic and isosmotic shrinkage on cellular functions were sharply different. In C11-MDCK and VSMC, hyperosmotic shrinkage completely inhibited Na(+),K(+)-ATPase and Na(+),P(i) cotransport. In contrast, in both types of cells isosmotic shrinkage slightly increased rather than suppressed Na(+),K(+)-ATPase and did not change Na(+),P(i) cotransport. In C11-MDCK cells, phosphorylation of JNK1/2 and Erk1/2 mitogen-activated protein kinases was augmented in hyperosmotically shrunken cells by ∼7- and 2-fold, respectively, but was not affected in cells subjected to isosmotic shrinkage. These results demonstrate that the data obtained in cells subjected to hyperosmotic shrinkage cannot be considered as sufficient proof implicating cell volume perturbations in the regulation of cellular functions under isosmotic conditions.


Subject(s)
Cell Size , Epithelial Cells/pathology , Kidney Tubules, Collecting/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Processing, Post-Translational , Sodium-Phosphate Cotransporter Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Dogs , Ion Transport , Kidney Tubules, Collecting/pathology , Kinetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Osmotic Pressure , Phosphorylation , Rats
5.
Can J Physiol Pharmacol ; 90(1): 83-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22188474

ABSTRACT

Na(+),K(+)-ATPase is a heterodimer consisting of catalytic α1-α4 and regulatory ß1-ß3 subunits. Recently, we reported that transfection with ouabain-resistant α1R-Na(+),K(+)-ATPase rescues renal epithelial C7-MDCK cells exclusively expressing the ouabain-sensitive α1S-isoform from the cytotoxic action of ouabain. To explore the role of α2 subunit in ion transport and cytotoxic action of ouabain, we compared the effect of ouabain on K(+) ((86)Rb) influx and the survival of ouabain-treated C7-MDCK cells stably transfected with α1R- and α2R-Na(+),K(+)-ATPase. α2R mRNA in transfected cells was ∼8-fold more abundant than α1R mRNA, whereas immunoreactive α2R protein content was 5-fold lower than endogenous α1S protein. A concentration of 10 µmol/L ouabain led to complete inhibition of (86)Rb influx both in mock- and α2R-transfected cells, whereas maximal inhibition of (86)Rb influx in α1R-transfectd cells was observed at 1000 µmol/L ouabain. In contrast to the massive death of mock- and α2R-transfected cells exposed to 3 µmol/L ouabain , α1R-cells survived after 24 h incubation with 1000 µmol/L ouabain. Thus, our results show that unlike α1R, the presence of α2R-Na(+),K(+)-ATPase subunit mRNA and immunoreactive protein does not contribute to Na(+)/K(+) pump activity, and does not rescue C7-MDCK cells from the cytotoxic action of ouabain. Our results also suggest that the lack of impact of transfected α2-Na(+),K(+)-ATPase on Na(+)/K(+) pump activity and cell survival can be attributed to the low efficiency of its translation and (or) delivery to the plasma membrane of renal epithelial cells.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/physiology , Ouabain/adverse effects , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , RNA Isoforms/genetics , Sodium-Potassium-Exchanging ATPase/biosynthesis , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Cell Death/drug effects , Cell Death/genetics , Cell Death/physiology , Cells, Cultured , Dogs , Ion Transport/drug effects , Ion Transport/genetics , Ion Transport/physiology , Isoenzymes/genetics , Kidney/drug effects , Kidney/metabolism , Kidney/physiology , Potassium/metabolism , Protein Biosynthesis/drug effects , Rubidium Radioisotopes/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Transfection/methods
6.
Pathophysiology ; 18(3): 207-13, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21247741

ABSTRACT

Numerous studies have demonstrated heightened Na(+)/Li(+) countertransport (NLCT) activity in erythrocytes of patients with essential hypertension or diabetic nephropathy. The same carrier also contributes to the therapeutic action of lithium salt, widely used in the treatment of psychiatric disorders. However, the molecular origin of NLCT remains unknown. This study examined the role of major ion transporters in NLCT by comparative analysis of its activity and that of ion transporters providing inwardly directed (86)Rb, (22)Na and (32)P fluxes. NLCT was below the detection limit in rat erythrocytes and ∼50-fold higher in rabbits compared to humans. Unlike NLCT, the activities of Na(+),K(+)-ATPase, Na(+),K(+),2Cl(-) cotransporter and anion exchanger were somewhat similar in the erythrocytes of these species, whereas Na(+),P(i) cotransport was in 1:2:6 proportion in rats, humans and rabbits, respectively. Loading of erythrocytes with Li(+) for NLCT measurement did not affect the activity of Na(+),P(i) cotransporter. Keeping in mind that NLCT is much higher in rabbits vs humans and rats, we compared the set of membrane proteins in these species using 2-dimensional gel electrophoresis. This approach revealed 174 common spots, whereas 132 proteins were detected only in human and rabbit erythrocyte membranes. Among these proteins, we found 17 spots whose expression was higher by more than 5-fold in rabbit compared to human erythrocytes. Thus, our results argue against the involvement of major ion transporters in NLCT. They also show that comparative proteomics is a potent tool to identify the molecular origin of this carrier.

7.
Am J Hypertens ; 23(3): 321-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20044742

ABSTRACT

BACKGROUND: Recent studies demonstrated a key role of ubiquitous isoform of Na+,K+,2Cl- co-transport (NKCC1) in regulation of myogenic tone and peripheral resistance. We examined the impact of race, gender, and plasma lipid on NKCC1 activity in French Canadians and African Americans with hypertension and dyslipidemia. METHODS: NKCC and passive erythrocyte membrane permeability to K+, measured as ouabain-resistant, bumetanide-sensitive, and (ouabain+bumetanide)-resistant 86Rb influx, respectively, were compared in 111 French-Canadian men, 107 French-Canadian women, 26 African-American men, and 45 African-American women with essential hypertension and dyslipidemia. RESULTS: The African-American men and women were 7 years younger and presented twofold decreased plasma triglycerides compared to their French-Canadian counterparts (P < 0.01) whereas body mass index (BMI), total cholesterol, low-density lipoprotein, and high-density lipoprotein (HDL) were not different. NKCC was respectively 50 and 38% lower in the African-American men and women than in the French Canadians (P < 0.005) without any differences in passive erythrocyte membrane permeability for K+. We did not observe any impact of age on NKCC in all groups under investigation, whereas plasma triglycerides correlated positively with the activity of this carrier in the French-Canadian men only. CONCLUSIONS: NKCC1 activity is lower in erythrocytes of African Americans with essential hypertension and dyslipidemia than in Caucasian counterparts. We suggest that decreased NKCC1 may contribute to the feature of the pathogenesis of salt-sensitive hypertension seen in African Americans.


Subject(s)
Black or African American , Dyslipidemias/blood , Erythrocytes/metabolism , Hypertension/blood , Sodium-Potassium-Chloride Symporters/metabolism , Adult , Aged , Aged, 80 and over , Animals , Body Mass Index , Female , Humans , Lipids/blood , Male , Middle Aged , Rats , Solute Carrier Family 12, Member 2 , White People , Young Adult
8.
Apoptosis ; 15(1): 55-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19949978

ABSTRACT

Mechanisms underlying the tissue-specific impact of cardiotonic steroids (CTS) on cell survival and death remain poorly understood. This study examines the role of Na(+),K(+)-ATPase alpha subunits in death of Madin-Darby canine kidney (MDCK) cells evoked by 24-h exposure to ouabain. MDCK cells expressing a variant of the alpha1 isoform, CTS-sensitive alpha1S, were stably transfected with a cDNA encoding CTS-resistant alpha1R-Na(+),K(+)-ATPase, whose expression was confirmed by RT-PCR. In mock-transfected and alpha1R-cells, maximal inhibition of (86)Rb influx was observed at 10 and 1000 muM ouabain, respectively, thus confirming high abundance of alpha1R-Na(+),K(+)-ATPase in these cells. Six-hour treatment of alpha1R-cells with 1000 muM ouabain led to the same elevation of the [Na(+)](i)/[K(+)](i) ratio that was detected in mock-transfected cells treated with 3 muM ouabain. However, in contrast to the massive death of mock-transfected cells exposed to 3 muM ouabain, alpha1R-cells survived after 24-h incubation with 1000 muM ouabain. Inversion of the [Na(+)](i)/[K(+)](i) ratio evoked by Na(+),K(+)-ATPase inhibition in K(+)-free medium did not affect survival of alpha1R-cells but increased their sensitivity to ouabain. Our results show that the alpha1R subunit rescues MDCK cells from the cytotoxic action of CTS independently of inhibition of Na(+),K(+)-ATPase-mediated Na(+) and K(+) fluxes and inversion of the [Na(+)](i)/[K(+)](i) ratio.


Subject(s)
Cardiac Glycosides/chemistry , Enzyme Inhibitors/chemistry , Epithelial Cells/enzymology , Kidney/enzymology , Ouabain/toxicity , Potassium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Animals , Cell Line , Dogs , Epithelial Cells/drug effects , Kidney/cytology , Kidney/drug effects , Protein Binding , Rats , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/genetics
9.
Apoptosis ; 14(11): 1266-73, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19784777

ABSTRACT

Recent studies demonstrate that cytotoxic actions of ouabain and other cardiotonic steroids (CTS) on renal epithelial cells (REC) are triggered by their interaction with the Na(+),K(+)-ATPase alpha-subunit but not the result of inhibition of Na(+),K(+)-ATPase-mediated ion fluxes and inversion of the [Na(+)](i)/[K(+)](i) ratio. This study examined the role of mitogen-activated protein kinases (MAPK) in the death of ouabain-treated REC. Exposure of C7-MDCK cells that resembled principal cells from canine kidney to 3 microM ouabain led to phosphorylation of p38 without significant impact on phosphorylation of ERK and JNK MAPK. Maximal increment of p38 phosphorylation was observed at 4 h followed by cell death at 12 h of ouabain addition. In contrast to ouabain, neither cell death nor p38 MAPK phosphorylation were affected by elevation of the [Na(+)](i)/[K(+)](i) ratio triggered by Na(+),K(+)-ATPase inhibition in K(+)-free medium. p38 phosphorylation was noted in all other cell types exhibiting death in the presence of ouabain, such as intercalated cells from canine kidney and human colon rectal carcinoma cells. We did not observe any action of ouabain on p38 phosphorylation in ouabain-resistant smooth muscle cells from rat aorta and endothelial cells from human umbilical vein. Both p38 phosphorylation and death of ouabain-treated C7-MDCK cells were suppressed by p38 inhibitor SB 202190 but were resistant to its inactive analogue SB 202474. Our results demonstrate that death of CTS-treated REC is triggered by Na (i) (+) ,K (i) (+) -independent activation of p38 MAPK.


Subject(s)
Epithelial Cells/pathology , Ouabain/pharmacology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cell Death/drug effects , Cell Line , Epithelial Cells/drug effects , Mitogen-Activated Protein Kinases/metabolism , Rats , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/physiology
10.
Apoptosis ; 13(5): 670-80, 2008 May.
Article in English | MEDLINE | ID: mdl-18392681

ABSTRACT

The mechanisms of cell death signaling triggered by cardiotonic steroids are poorly understood. Based on massive detachment of ouabain-treated Madin-Darby canine kidney (MDCK) cells, it may be proposed that the cytotoxic action of these compounds is mediated by anoikis, i.e. a particular mode of death occurring in cells lacking cell-to-extracellular matrix interactions. We tested this hypothesis. Six hour incubation of MDCK cells with ouabain, marinobufagenin or K+-free medium almost completely blocked Na+,K+-ATPase, increased Na (i) + content by approximately 10-fold and suppressed cell attachment to regular-plastic-plates by up to 5-fold. In contrast, the death of attached cells was observed after 24-h incubation with ouabain but not in the presence of marinobufagenin or K+-free medium. Cells treated with ouabain and undergoing anoikis on ultra-low attachment plates exhibited different cell volume behaviour, i.e. swelling and shrinkage, respectively. The pan-caspase inhibitor z-VAD.fmk and the protein kinase C activator PMA rescued MDCK cells from anoikis but did not influence the survival of ouabain-treated cells, whereas medium acidification from pH 7.2 to 6.7 almost completely abolished the cytotoxic action of ouabain, but did not significantly affect anoikis. Our results show that the Na (i) + ,K (i) + -independent mode of MDCK cell death evoked by ouabain is not mediated by anoikis.


Subject(s)
Anoikis/physiology , Cell Death/drug effects , Kidney/cytology , Ouabain/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Anoikis/drug effects , Bufanolides/pharmacology , Caspase 3/metabolism , Caspase Inhibitors , Cell Survival/drug effects , Cells, Cultured , Chromatin/physiology , Dogs , Enzyme Activation , Hydrogen-Ion Concentration , Kidney/drug effects , Microscopy, Phase-Contrast , Protein Kinase C/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
11.
Cell Physiol Biochem ; 21(4): 315-24, 2008.
Article in English | MEDLINE | ID: mdl-18441520

ABSTRACT

Recently, we reported that the death of ouabain-treated C7-MDCK cells resembling principal cells from collecting ducts of the Madin-Darby canine kidney (MDCK) is caused by ouabain interaction with Na+,K+-ATPase but is not mediated by inversion of the [Na+](i)/[K+](i) ratio. The mechanism of this intriguing phenomenon remains unknown. We therefore examined the action of ouabain on serine/threonine phosphoproteins as possible intermediates of cell death signaling. The death of ouabain-treated C7-MDCK cells proceeded by altered phosphorylation of the RRXS*/T*-motif in 4 proteins with Mr from 80 to 25 kDa. Similarly to cell death, inversion of the [Na+](i)/[K+](i) ratio evoked by Na+,K+-ATPase inhibition in K+-free medium did not affect the phosphorylation of RRXS*/T*-proteins but increased their sensitivity to ouabain. The action of ouabain was preserved in the presence of activators of protein kinases A (forskolin), G (sodium nitroprusside) and C (PMA) as well as inhibitors of protein kinase C (Go 6983, Go 6976) and serine-threonine phosphatases (okadaic acid). Phosphorylation of RRXS*/T*-proteins was also noted in ouabain-sensitive C11-MDCK cells resembling intercalated cells from collecting ducts, but was absent in ouabain-resistant smooth muscle cells from the rat aorta. Our results show that altered phosphorylation of RRXS*/T*-proteins in ouabain-treated C7-MDCK cells is mediated by its interaction with Na+,K+-ATPase but is not caused by inversion of the [Na+](i)/[K+](i) ratio. The molecular origin of serine-threonine kinases and/or phosphatases involved in phosphorylation of ouabain-sensitive proteins and their role in cell death signaling should be examined further.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , Kidney/cytology , Ouabain/pharmacology , Potassium/metabolism , Sodium/metabolism , Amino Acid Motifs , Animals , Cells, Cultured , Culture Media, Conditioned , Dogs , Male , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Inbred WKY , Substrate Specificity
12.
Purinergic Signal ; 4(2): 183-91, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18368525

ABSTRACT

Previously, we observed that sustained activation of P2Y1 leads to inhibition of Na⁺,K⁺,Cl⁻ cotransport (NKCC) in C11 cells resembling intercalated cells from collecting ducts of the Madin-Darby canine kidney. This study examined the role of stress-activated protein kinases (SAPK) in NKCC inhibition triggered by purinergic receptors. Treatment of C11 cells with ATP led to sustained phosphorylation of SAPK such as JNK and p38. Activation of these kinases also occurred in anisomycin-treated cells. Surprisingly, we observed that compounds SP600125 and SB202190, known as potent inhibitors of JNK and p38 in cell-free systems, activated rather than inhibited phosphorylation of the kinases in C11 cells. Importantly, similarly to ATP, all the above-listed activators of JNK and p38 phosphorylation inhibited NKCC. Thus, our results suggest that activation of JNK and/or p38 contributes to NKCC suppression detected in intercalated-like cells from distal tubules after their exposure to P2Y1 agonists.

13.
Pflugers Arch ; 455(4): 711-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17622553

ABSTRACT

The cytotoxic effect of long-term exposure of renal epithelial cells to ouabain and other cardiotonic steroids (CTS) is mediated by the interaction of these compounds with Na(+),K(+)-ATPase but is independent of the inhibition of Na(+),K(+)-ATPase-mediated ion fluxes. Sustained application of CTS also leads to Na(+),K(+)-ATPase endocytosis and its translocation into the nuclei that might trigger the cell death machinery via the regulation of gene expression. This study examines the role of Na(+),K(+)-ATPase internalization and de novo gene expression in the death of ouabain-treated C7-Madin-Darby canine kidney (MDCK) cells derived from distal tubules of the MDCK. In these cells, 6-h exposure to 3 microM ouabain led to the internalization of approximately 50% of plasmalemmal Na(+),K(+)-ATPase. Prolonged incubation in a K(+)-free medium abolished ouabain-induced Na(+),K(+)-ATPase internalization but did not affect the cytotoxic action of ouabain seen after 18-h incubation. Previously, it was shown that CTS-induced Na(+),K(+)-ATPase internalization is mediated by its interaction with Src within caveolae. Neither caveolae damage by cholesterol depletion with methyl-beta-cyclodextrin nor Src inhibition with 4-amino-5(4-chlorophenyl)-7-(t-butyl)pyrazol[3,4-d]pyridine affected the death of ouabain-treated C7-MDCK cells. Actinomycin D at the 0.1-microg/ml concentration almost completely abolished ribonucleic acid synthesis but did not protect C7-MDCK cells from the cytotoxic action of ouabain. Our results show that neither Na(+),K(+)-ATPase endocytosis nor de novo gene expression contributes to Na(+)(i), K(+)(i)-independent cell death signaling evoked by prolonged exposure to CTS.


Subject(s)
Cardiotonic Agents/toxicity , Enzyme Inhibitors/toxicity , Epithelial Cells/drug effects , Kidney/drug effects , Ouabain/toxicity , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Animals , Caveolae/drug effects , Caveolae/metabolism , Cell Death/drug effects , Cell Line , Cholesterol/deficiency , Dactinomycin/pharmacology , Dogs , Endocytosis/drug effects , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression/drug effects , Kidney/enzymology , Kidney/metabolism , Kidney/pathology , Nucleic Acid Synthesis Inhibitors/pharmacology , Potassium/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Time Factors , beta-Cyclodextrins/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
14.
FEBS J ; 274(14): 3557-3567, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17565602

ABSTRACT

In vascular smooth muscle cells and several other cell types, inhibition of Na(+)/K(+)-ATPase leads to the expression of early response genes, including c-Fos. We designed this study to examine whether or not a putative Na(+) (i)/K(+) (i)-sensitive element is located within the c-Fos 5'-UTR from - 650 to + 103 containing all known response elements activated by 'classic' stimuli, such as growth factors and Ca(2+) (i)-raising compounds. In HeLa cells, the highest increment of c-Fos mRNA content was noted after 6 h of Na(+)/K(+)-ATPase inhibition with ouabain that was abolished by actinomycin D, an inhibitor of RNA synthesis. c-Fos protein accumulation in ouabain-treated cells correlated with a gain of Na(+) (i) and loss of K(+) (i). Augmented c-Fos expression was also observed under inhibition of Na(+)/K(+)-ATPase in K(+)-free medium and in the presence of the Na(+) ionophore monensin. The effect of ouabain on c-Fos expression was sharply attenuated under dissipation of the transmembrane Na(+) gradient, but was preserved in the presence of Ca(2+) chelators and the extracellular regulated kinase inhibitor PD98059, thus indicating an Na(+) (i)-mediated, Ca(2+) (i)- and extracellular regulated kinase-independent mechanism of gene expression. In contrast to massive c-Fos expression, we failed to detect any effect of ouabain on accumulation of luciferase driven by the c-Fos 5'-UTR. Negative results were also obtained in ouabain-treated vascular smooth muscle cells and C11 Madin-Darby canine kidney cells possessing augmented c-Fos expression. Our results reveal that Na(+) (i)-induced c-Fos expression is not mediated by the 5'-UTR containing transcriptional elements activated by growth factors and other 'classic stimuli'.


Subject(s)
Gene Expression Regulation/drug effects , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Sodium/metabolism , Sodium/pharmacology , Transcription, Genetic/genetics , Animals , Calcium/metabolism , Cell Line , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, Reporter/genetics , Humans , Mice , Ouabain/pharmacology , RNA/biosynthesis , RNA, Messenger/genetics , Sensitivity and Specificity , Serum Response Element , Signal Transduction , Transcription, Genetic/drug effects
15.
J Biol Chem ; 281(42): 31317-25, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16916802

ABSTRACT

In C11-MDCK cells, which resemble intercalated cells from collecting ducts of the canine kidney, P2Y agonists promote transient activation of the Na+,K+,Cl- cotransporter (NKCC), followed by its sustained inhibition. We designed this study to identify P2Y receptor subtypes involved in dual regulation of this carrier. Real time polymerase chain reaction analysis demonstrated that C11-MDCK cells express abundant P2Y1 and P2Y2 mRNA compared with that of other P2Y receptor subtypes. The rank order of potency of agents (ATP approximately UTP >> 2-(methylthio)-ATP (2MeSATP); adenosine 5'-[beta-thio]diphosphate (ADPbetaS) inactive) indicated that P2Y2 rather than P2Y1 receptors mediate a 3-4-fold activation of NKCC within the first 5-10 min of nucleotide addition. NKCC activation in ATP-treated cells was abolished by the intracellular calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid, calmodulin (CaM) antagonists trifluoroperazine and W-7, and KN-62, an inhibitor of Ca2+/CaM-dependent protein kinase II. By contrast with the transient activation, 30-min incubation with nucleotides produced up to 4-5-fold inhibition of NKCC, and this inhibition exhibited a rank order of potency (2MeSATP > ADPbetaS > ATP >> UTP) typical of P2Y1 receptors. Unlike the early response, delayed inhibition of NKCC occurred in 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-loaded cells and was completely abolished by the P2Y1 antagonists MRS2179 and MRS2500. Transient activation and delayed inhibition of NKCC in C11 cell monolayers were observed after the addition of ATP to mucosal and serosal solutions, respectively. NKCC inhibition triggered by basolateral application of ADPbetaS was abolished by MRS2500. Our results thus show that transient activation and delayed inhibition of NKCC in ATP-treated C11-MDCK cells is mediated by Ca2+/CaM-dependent protein kinase II- and Ca2+-independent signaling triggered by apical P2Y2 and basolateral P2Y1 receptors, respectively.


Subject(s)
Adenosine Triphosphate/pharmacology , Calmodulin/metabolism , Receptors, Purinergic P2/physiology , Sodium-Potassium-Chloride Symporters/physiology , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Biological Transport , Chlorine/chemistry , Deoxyadenine Nucleotides/pharmacology , Dogs , Enzyme Activation , Gene Expression Regulation , Kinetics , Potassium/metabolism , Receptors, Purinergic P2/metabolism , Solute Carrier Family 12, Member 2
16.
Pflugers Arch ; 451(4): 569-78, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16052351

ABSTRACT

The signaling cascade resulting in the death of several types of cells treated with ouabain or other cardiotonic steroids (CTS) remains poorly understood. Recently, we observed that ouabain kills epithelial and endothelial cells via its interaction with Na(+), K(+) -ATPase, but independently of inhibition of Na(+), K(+) -ATPase-mediated ion fluxes and inversion of the [Na(+)](i)/[K(+)](i) ratio. Here, we report that the death of ouabain-treated epithelial cells from the Madin-Darby canine kidney (C7-MDCK) and endothelial cells from porcine aortae is suppressed by acidification of medium from pH 7.4 to 7.0, i.e. under conditions when pH(i) was decreased from approximately 7.2 to 6.9. The rescue of ouabain-treated C7-MDCK cells was also detected under selective intracellular acidification caused by inhibition of Na(+)/H(+) exchanger. In these cells, neither Na(+), K(+) pump activity nor [(3)H]-ouabain binding was significantly affected by modest acidification. The death of ouabain-treated cells was independent of inhibition of RNA and protein synthesis with actinomycin D and cycloheximide. In contrast, both compounds sharply attenuated the protective action of acidified medium. Thus, our results show that very modest intracellular acidification is sufficient to inhibit the Na(+) (i)/K(+) (i)-independent death signal triggered in epithelial and endothelial cells by CTS. They also suggest that the protective action of acidification is mediated by de novo expression of genes involved in inhibition of the cell death machinery.


Subject(s)
Enzyme Inhibitors/pharmacology , Ouabain/pharmacology , Signal Transduction/drug effects , Animals , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Dogs , Hydrogen-Ion Concentration
17.
Pathophysiology ; 12(2): 125-35, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16023561

ABSTRACT

Previously, we reported that ouabain and other cardiotonic steroids (CTS) kill renal epithelial and vascular endothelial cells via their interaction with the Na+,K+-ATPase alpha-subunit, but independently of elevation of the [Na+]i/[K+]i ratio. In distinct cell types, side-by-side with inhibition of Na+,K+-ATPase-mediated ion fluxes, CTS trigger [Ca2+]i oscillation, activation of Ras, mitogen-activated protein kinases (MAPK), phosphoinositide-3 kinase (PI3K), and protein kinase C as well as the production of reactive oxygen species and cytoskeleton reorganization. This study examined the potential involvement of the above-listed intermediates in death signaling triggered by ouabain in C7-Madin-Darby canine kidney cells. In these cells, twofold decreased staining with dimethylthiazol diphenyltetrazolium (MTT) and detachment of up to 80% of dead cells were detected in 6 and 24 h of ouabain addition, respectively. We did not observe any effect of extra- (EGTA) and intracellular (BAPTA) Ca2+-chelators, [Ca2+]i-raising compounds (thapsigargin, ATP), inhibitors of Ras signaling (alpha-hydroxyfarnesyl-sulphosphoric acid), PI3K (wortmannin), MAPK ERK1/2 kinase (PD98059), tyrosine kinases (genistein) as well as activators (4beta-PMA, 8-Br-cAMP, 8-Br-cGMP, forskolin) and inhibitors (calphostin) of serine-threonine kinases on MTT staining and death of ouabain-treated cells. Ouabain did not affect cellular redox state and the production of superoxide anion and hydroperoxide. Neither N-acetylcysteine nor reduced gluthatione suppressed the death of ouabain-treated cells. Thus, our results show that none of the above-listed signaling systems plays a major role in the development of Nai+,Ki+-independent death machinery triggered by CTS interaction with the Na+,K+-ATPase alpha-subunit.

18.
J Biol Chem ; 280(1): 832-9, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15494417

ABSTRACT

Recently, we reported that ouabain kills renal epithelial and vascular endothelial cells independently of elevation of the [Na(+)](i)/[K(+)](i) ratio. These observations raised the possibility of finding cardiotonic steroids (CTS) that inhibit the Na(+),K(+) pump without attenuating cell survival and vice versa. To test this hypothesis, we compared CTS action on Na(+),K(+) pump, [Na(+)](i) content, and survival of Madin-Darby canine kidney cells. At a concentration of 1 microM, ouabain and other tested cardenolides, as well as bufadienolides such as bufalin, cinobufagin, cinobufotalin, and telobufotoxin, led to approximately 10-fold inhibition of the Na(+),K(+) pump, a 2-3-fold decrease in staining with dimethylthiazol-diphenyltetrazolium (MTT), and massive death indicated by detachment of approximately 80% of cells and caspase-3 activation. In contrast, Na(+),K(+) pump inhibition and elevation of [Na(+)](i) seen in the presence of 3 microM marinobufagenin (MBG) and marinobufotoxin did not affect MTT staining and cell survival. Inhibition of the Na(+),Rb(+) pump in K(+)-free medium was not accompanied by a decline of MTT staining and cell detachment but increased sensitivity to CTS. In K(+)-free medium, half-maximal inhibition of (86)Rb influx was observed in the presence of 0.04 microM ouabain and 0.1 microM MBG, whereas half-maximal detachment and decline of MTT staining were detected at 0.03 and 0.004 microM of ouabain versus 10 and 3 microM of MBG, respectively. Both ouabain binding and ouabain-induced [Na(+)](i),[K(+)](i)-independent signaling were suppressed in the presence of MBG. Thus, our results show that CTS exhibit distinctly different potency in Na(+),K(+) pump inhibition and triggering of [Na(+)](i)/[K(+)](i)-independent signaling, including cell death.


Subject(s)
Cardiac Glycosides/pharmacology , Potassium/metabolism , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium/metabolism , Animals , Cell Line , Cell Survival/drug effects , Dogs
SELECTION OF CITATIONS
SEARCH DETAIL
...