Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cytometry B Clin Cytom ; 102(3): 220-227, 2022 05.
Article in English | MEDLINE | ID: mdl-35253974

ABSTRACT

BACKGROUND: A key step in clinical flow cytometry data analysis is gating, which involves the identification of cell populations. The process of gating produces a set of reportable results, which are typically described by gating definitions. The non-standardized, non-interpreted nature of gating definitions represents a hurdle for data interpretation and data sharing across and within organizations. Interpreting and standardizing gating definitions for subsequent analysis of gating results requires a curation effort from experts. Machine learning approaches have the potential to help in this process by predicting expert annotations associated with gating definitions. METHODS: We created a gold-standard dataset by manually annotating thousands of gating definitions with cell type and functional marker annotations. We used this dataset to train and test a machine learning pipeline able to predict standard cell types and functional marker genes associated with gating definitions. RESULTS: The machine learning pipeline predicted annotations with high accuracy for both cell types and functional marker genes. Accuracy was lower for gating definitions from assays belonging to laboratories from which limited or no prior data was available in the training. Manual error review ensured that resulting predicted annotations could be reused subsequently as additional gold-standard training data. CONCLUSIONS: Machine learning methods are able to consistently predict annotations associated with gating definitions from flow cytometry assays. However, a hybrid automatic and manual annotation workflow would be recommended to achieve optimal results.


Subject(s)
Machine Learning , Flow Cytometry , Humans , Workflow
2.
Biochem Biophys Rep ; 12: 220-227, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29159314

ABSTRACT

OBJECTIVES: This study examined the dose-dependent actions of hydrogen sulfide donor sodium hydrosulphide (NaHS) on isometric contractions and ion transport in rat aorta smooth muscle cells (SMC). METHODS: Isometric contraction was measured in ring aortas segments from male Wistar rats. Activity of Na+/K+-pump and Na+,K+,2Cl-cotransport was measured in cultured endothelial and smooth muscle cells from the rat aorta as ouabain-sensitive and ouabain-resistant, bumetanide-sensitive components of the 86Rb influx, respectively. RESULTS: NaHS exhibited the bimodal action on contractions triggered by modest depolarization ([K+]o=30 mM). At 10-4 M, NaHS augmented contractions of intact and endothelium-denuded strips by ~ 15% and 25%, respectively, whereas at concentration of 10-3 M it decreased contractile responses by more than two-fold. Contractions evoked by 10-4 M NaHS were completely abolished by bumetanide, a potent inhibitor of Na+,K+,2Cl-cotransport, whereas the inhibition seen at 10-3 M NaHS was suppressed in the presence of K+ channel blocker TEA. In cultured SMC, 5×10-5 M NaHS increased Na+,K+,2Cl- - cotransport without any effect on the activity of this carrier in endothelial cells. In depolarized SMC, 45Ca influx was enhanced in the presence of 10-4 M NaHS and suppressed under elevation of [NaHS] up to 10-3 M. 45Ca influx triggered by 10-4 M NaHS was abolished by bumetanide and L-type Ca2+ channel blocker nicardipine. CONCLUSIONS: Our results strongly suggest that contractions of rat aortic rings triggered by low doses of NaHS are mediated by activation of Na+,K+,2Cl-cotransport and Ca2+ influx via L-type channels.

3.
Cell Calcium ; 68: 5-13, 2017 12.
Article in English | MEDLINE | ID: mdl-29129208

ABSTRACT

In rat vascular smooth muscle cells (RVSMC), 3-h Na+,K+-ATPase inhibition by ouabain or in K+-free medium resulted in the inversion of the [Na+]i/[K+]i ratio and elevation up to 7-fold the content of Egr1, Atf3, Nr4a1 and Ptgs2 mRNAs. Ouabain increased the rate of 45Ca2+ influx by 2-fold that was abolished by L-type voltage-gated Ca2+ channel blocker nicardipine, but it was resistant to Na+/Ca2+ exchanger inhibitor KB-R7943. To study the role of Ca2+-mediated signaling in the expression of Na+i/K+i-sensitive genes we used intracellular Ca2+ chelator BAPTA and incubated RVSMC in Ca2+-free medium. The elevation of Nr4a1 and Ptgs2 expression triggered by ouabain was diminished in Ca2+-depeleted cells as well as in the presence of nicardipine and calmodulin antagonists A-7 and W-7. Ptgs2 expression was also suppressed by inhibitor of Ca2+/calmodulin-dependent protein kinase (CaMKII) KN-93 whereas increment of Nr4a1 content triggered by ouabain was attenuated by inhibitor of Ca2+/calmodulin-dependent protein phosphatase (calcineurin, CaN) cyclosporin A. Neither Ca2+ depletion nor above listed compounds had any impact on the augmented expression of Egr1 and Atf3 in ouabain-treated RVSMC. Our results strongly suggest that dissipation of transmembrane gradient of monovalent cations increases Ptgs2 and Nr4a1 transcription via augment Ca2+ influx through L-type Ca2+ channels that, in turn, leads to CaMKII-mediated phosphorylation of CREB and calcineurin-mediated dephosphorylation of NFAT, respectively. Additional experiments should be performed to identify intermediates of Na+i,K+i-mediated Ca2+-independent excitation-transcription coupling involved the regulation of Egr1 and Atf3 expression.


Subject(s)
Calcium/metabolism , Gene Expression Regulation , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Transcription, Genetic , Animals , Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Gene Expression Regulation/drug effects , Hydrogen-Ion Concentration , Kinetics , Male , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Nicardipine/pharmacology , Ouabain/pharmacology , Potassium/metabolism , Rats, Wistar , Signal Transduction/drug effects , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Thiourea/analogs & derivatives , Thiourea/pharmacology , Transcription, Genetic/drug effects
4.
Sci Rep ; 7: 45403, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28345607

ABSTRACT

Recent studies demonstrated that in addition to Na+,K+-ATPase inhibition cardiotonic steroids (CTSs) affect diverse intracellular signaling pathways. This study examines the relative impact of [Na+]i/[K+]i-mediated and -independent signaling in transcriptomic changes triggered by the endogenous CTSs ouabain and marinobufagenin (MBG) in human umbilical vein endothelial cells (HUVEC). We noted that prolongation of incubation increased the apparent affinity for ouabain estimated by the loss of [K+]i and gain of [Na+]i. Six hour exposure of HUVEC to 100 and 3,000 nM ouabain resulted in elevation of the [Na+]i/[K+]i ratio by ~15 and 80-fold and differential expression of 258 and 2185 transcripts, respectively. Neither [Na+]i/[K+]i ratio nor transcriptome were affected by 6-h incubation with 30 nM ouabain. The 96-h incubation with 3 nM ouabain or 30 nM MBG elevated the [Na+]i/[K+]i ratio by ~14 and 3-fold and led to differential expression of 880 and 484 transcripts, respectively. These parameters were not changed after 96-h incubation with 1 nM ouabain or 10 nM MBG. Thus, our results demonstrate that elevation of the [Na+]i/[K+]i ratio is an obligatory step for transcriptomic changes evoked by CTS in HUVEC. The molecular origin of upstream [Na+]i/[K+]i sensors involved in transcription regulation should be identified in forthcoming studies.


Subject(s)
Cardiac Glycosides/pharmacology , Cardiotonic Agents/pharmacology , Ions/metabolism , Potassium/metabolism , Sodium/metabolism , Transcriptome/drug effects , Bufanolides/pharmacology , Cell Line , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Ouabain/pharmacology , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Transcription, Genetic/drug effects
5.
Am J Physiol Lung Cell Mol Physiol ; 310(9): L815-23, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26851261

ABSTRACT

Myofibroblast differentiation is a key process in pathogenesis of fibrotic diseases. Cardiac glycosides (ouabain, digoxin) inhibit Na(+)-K(+)-ATPase, resulting in increased intracellular [Na(+)]-to-[K(+)] ratio in cells. Microarray analysis suggested that increased intracellular [Na(+)]/[K(+)] ratio may promote the expression of cyclooxygenase-2 (COX-2), a critical enzyme in the synthesis of prostaglandins. Given antifibrotic effects of prostaglandins through activation of protein kinase A (PKA), we examined if cardiac glycosides stimulate COX-2 expression in human lung fibroblasts and how they affect myofibroblast differentiation. Ouabain stimulated a profound COX-2 expression and a sustained PKA activation, which was blocked by COX-2 inhibitor or by COX-2 knockdown. Ouabain-induced COX-2 expression and PKA activation were abolished by the inhibitor of the Na(+)/Ca(2+) exchanger, KB-R4943. Ouabain inhibited transforming growth factor-ß (TGF-ß)-induced Rho activation, stress fiber formation, serum response factor activation, and the expression of smooth muscle α-actin, collagen-1, and fibronectin. These effects were recapitulated by an increase in intracellular [Na(+)]/[K(+)] ratio through the treatment of cells with K(+)-free medium or with digoxin. Although inhibition of COX-2 or of the Na(+)/Ca(2+) exchanger blocked ouabain-induced PKA activation, this failed to reverse the inhibition of TGF-ß-induced Rho activation or myofibroblast differentiation by ouabain. Together, these data demonstrate that ouabain, through the increase in intracellular [Na(+)]/[K(+)] ratio, drives the induction of COX-2 expression and PKA activation, which is accompanied by a decreased Rho activation and myofibroblast differentiation in response to TGF-ß. However, COX-2 expression and PKA activation are not sufficient for inhibition of the fibrotic effects of TGF-ß by ouabain, suggesting that additional mechanisms must exist.


Subject(s)
Cardiac Glycosides/pharmacology , Cell Differentiation , Digoxin/pharmacology , Myofibroblasts/physiology , Ouabain/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Enzyme Activation , Gene Expression , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Myofibroblasts/drug effects
6.
Cell Calcium ; 58(3): 317-24, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26183762

ABSTRACT

Previously, we reported that Ca(2+) depletion increased permeability of the plasma membrane for Na(+). This study examined the relative impact of [Na(+)]i/[K(+)]i-mediated signaling on transcriptomic changes in cultured vascular smooth muscle cells from rat aorta (VSMC) subjected to Ca(2+)-depletion by extra-(EGTA) and intracellular (BAPTA-AM) Ca(2+) chelators. Na(+),K(+)-ATPase inhibition in K(+)-free medium during 3 h led to elevation of [Na(+)]i and attenuation of [K(+)]i by ∼7- and 10-fold, whereas Ca(2+)-depletion resulted in alteration of these parameters by ∼3- and 2-fold, respectively. Augmented VSMC permeability for Na(+) and elevation of the [Na(+)]i/[K(+)]i ratio was triggered by addition to Ca(2+)-free medium 50 µM EGTA and was not affected by 10 µM BAPTA-AM. Na(+),K(+)-ATPase inhibition and Ca(2+)-depletion changed expression of 3677 and 4610 mRNA transcripts, respectively. We found highly significant (p<10(-12)) positive (R(2)>0.51) correlation between levels of expression of 2071 transcripts whose expression was affected by both stimuli. Among genes whose expression in Ca(2+)-depleted cells was augmented by more than 7-fold we noted cyclic AMP-dependent transcription factor Atf3, early growth response protein Egr1 and nuclear receptor subfamily 4, group A member Nr4a1. Dissipation of transmembrane gradients of monovalent cations in high-K(+), low-Na(+)-medium abolished the increments of the [Na(+)]i/[K(+)]i ratio as well as the augmented expression of these genes triggered by incubation of VSMC in EGTA containing medium. Thus, our results demonstrate, for the first time, that robust transcriptomic changes triggered by Ca(2+)-depletion in the presence of extracellular Ca(2+)-chelators are at least partially mediated by elevation of the [Na(+)]i/[K(+)]i ratio and activation of Ca(2+)i-independent, [Na(+)]i/[K(+)]i-mediated mechanism of excitation-transcription coupling. These results shad a new light on analysis of data obtained in cells subjected to long-term exposure to Ca(2+) chelators.


Subject(s)
Calcium Signaling , Calcium/metabolism , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Muscle, Smooth, Vascular/physiology , Potassium/metabolism , Sodium/metabolism , Transcriptome , Animals , Calcium/deficiency , Cells, Cultured , Egtazic Acid/pharmacology , Gene Expression Profiling , Rats , Transcriptome/drug effects
7.
Genes Dis ; 2(2): 186-196, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26114157

ABSTRACT

This review summarizes the data on the functional significance of ubiquitous (NKCC1) and renal-specific (NKCC2) isoforms of electroneutral sodium, potassium and chloride cotransporters. These carriers contribute to the pathogenesis of hypertension via regulation of intracellular chloride concentration in vascular smooth muscle and neuronal cells and via sensing chloride concentration in the renal tubular fluid, respectively. Both NKCC1 and NKCC2 are inhibited by furosemide and other high-ceiling diuretics widely used for attenuation of extracellular fluid volume. However, the chronic usage of these compounds for the treatment of hypertension and other volume-expanded disorders may have diverse side-effects due to suppression of myogenic response in microcirculatory beds.

8.
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
9.
J Biol Chem ; 289(11): 7505-13, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24492608

ABSTRACT

Myofibroblast differentiation is a key process in the pathogenesis of fibrotic disease. We have shown previously that differentiation of myofibroblasts is regulated by microtubule polymerization state. In this work, we examined the potential antifibrotic effects of the antitussive drug, noscapine, recently found to bind microtubules and affect microtubule dynamics. Noscapine inhibited TGF-ß-induced differentiation of cultured human lung fibroblasts (HLFs). Therapeutic noscapine treatment resulted in a significant attenuation of pulmonary fibrosis in the bleomycin model of the disease. Noscapine did not affect gross microtubule content in HLFs, but inhibited TGF-ß-induced stress fiber formation and activation of serum response factor without affecting Smad signaling. Furthermore, noscapine stimulated a rapid and profound activation of protein kinase A (PKA), which mediated the antifibrotic effect of noscapine in HLFs, as assessed with the PKA inhibitor, PKI. In contrast, noscapine did not activate PKA in human bronchial or alveolar epithelial cells. Finally, activation of PKA and the antifibrotic effect of noscapine in HLFs were blocked by the EP2 prostaglandin E2 receptor antagonist, PF-04418948, but not by the antagonists of EP4, prostaglandin D2, or prostacyclin receptors. Together, we demonstrate for the first time the antifibrotic effect of noscapine in vitro and in vivo, and we describe a novel mechanism of noscapine action through EP2 prostaglandin E2 receptor-mediated activation of PKA in pulmonary fibroblasts.


Subject(s)
Antitussive Agents/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Noscapine/pharmacology , Pulmonary Fibrosis/drug therapy , Receptors, Prostaglandin E/metabolism , Animals , Antineoplastic Agents/pharmacology , Bleomycin/pharmacology , Cell Line, Tumor , DNA/metabolism , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation , Humans , Hydroxyproline/chemistry , Luciferases/metabolism , Lung/metabolism , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Myofibroblasts/cytology , Neoplasms/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology
10.
Hypertension ; 62(4): 731-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23959560

ABSTRACT

Elevation of blood pressure with age is one of the hallmarks of hypertension in both males and females. This study examined transcriptomic profiles in the kidney of 12-, 40-, and 80-week-old spontaneously hypertensive rats and 4 recombinant inbred strains in search for functional genetic elements supporting temporal dynamics of blood pressure elevation. We found that both in males and females of spontaneously hypertensive rats and hypertensive recombinant inbred strains age-dependent blood pressure increment was accompanied by 50% heightened expression of epithelial sodium channel ß- and γ-subunits. Epithelial sodium channel subunit expression correlated positively with blood pressure but correlated negatively with renin expression. Increased epithelial sodium channel activity was observed in cultured epithelial cells isolated from the kidney medulla of 80-week-old spontaneously hypertensive rats but not in age-matched normotensive Wistar Kyoto. This difference remained evident after 24-hour treatment with aldosterone. 22Na uptake in the perfused kidney medulla was increased whereas the urinary Na/K ratio was decreased in old spontaneously hypertensive rats compared with normotensive controls. The difference was eliminated by the administration of epithelial sodium channel inhibitor benzamil. Observations in recombinant inbred strains representing various mixtures of parental hypertensive and normotensive genomes suggest that Scnn1g and Scnn1b genes themselves are not implicated in heightened expression and that the increased expression is neither secondary nor required for a partial elevation of blood pressure in contrast to spontaneously hypertensive rats. We suggest that spontaneously hypertensive rats display an intact negative feed-back between renin-angiotensin-system and epithelial Na channel activity whose upregulated expression is supported by a yet unknown mechanism.


Subject(s)
Aging/metabolism , Blood Pressure/physiology , Epithelial Sodium Channels/metabolism , Hypertension/metabolism , Kidney/metabolism , Aging/genetics , Animals , Epithelial Sodium Channels/genetics , Female , Hypertension/genetics , Hypertension/physiopathology , Kidney/physiopathology , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Renin-Angiotensin System/genetics , Up-Regulation
11.
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
12.
Ann Med ; 44 Suppl 1: S111-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22713139

ABSTRACT

High-ceiling diuretics (HCD), known potent inhibitors of housekeeping Na(+),K(+),2Cl cotransporter (NKCC1) and renal-specific NKCC2, decrease [Cl(-)](i), hyperpolarize vascular smooth muscle cells (VSMC), and suppress contractions evoked by modest depolarization, phenylephrine, angiotensin II, and UTP. These actions are absent in nkcc1 (/) knock-out mice, indicating that HCD interact with NKCC1 rather than with other potential targets. These findings also suggest that VSMC-specific inhibitors of NKCC1 may be considered potential pharmacological therapeutic tools in treatment of hypertension. It should be underlined that side by side with attenuation of peripheral resistance and systemic blood pressure, HCD blocked myogenic tone (MT) in renal afferent arterioles. Keeping this in mind, attenuation of MT might be a mechanism underlying the prevalence of end-stage renal disease documented in hypertensive African-Americans with decreased NKCC1 activity and in hypertensive patients subjected to chronic HCD treatment. The role of NKCC1-mediated MT in protection of the brain, heart, and other encapsulated organs deserves further investigation.


Subject(s)
Hypertension/metabolism , Muscle Contraction , Muscle, Smooth, Vascular/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Blood Pressure , Humans , Hypertension/etiology , Hypertension/physiopathology , Muscle, Smooth, Vascular/physiopathology , Solute Carrier Family 12, Member 2 , Vasoconstriction
13.
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
14.
Apoptosis ; 17(5): 429-38, 2012 May.
Article in English | MEDLINE | ID: mdl-22249286

ABSTRACT

Contrasting cell volume behaviours (swelling vs. shrinkage) are considered as criteria to distinguish necrosis from apoptosis. In this study, we employed a time-lapse, dual-image surface reconstruction technique to assess the volume of single vascular smooth muscle cells transfected with E1A-adenoviral protein (E1A-VSMC) and undergoing rapid apoptosis in the absence of growth factors or in the presence of staurosporine. After 30- to 60-min lag-phase, serum-deprived E1A-VSMC volume was increased by ~40%, which preceded maximal increments of caspase-3 activity and chromatin cleavage. Swollen cells underwent rapid apoptotic collapse, documented by plasma membrane budding, and terminated in 10-15 min by the formation of numerous apoptotic bodies. Suppression of apoptosis by inhibition of Na(+),K(+)-ATPase and activation of cAMP signalling with ouabain and forskolin, respectively, completely abolished the swelling of serum-deprived E1A-VSMC. In contrast to serum deprivation, apoptotic collapse of staurosporine-treated E1A-VSMC preceded attenuation of their volume by ~30%. Neither transient hyposmotic swelling nor isosmtotic shrinkage triggered apoptosis. Our results show that cell shrinkage can not be considered as ubiquitous hallmark of apoptosis. The involvement of stimulus-specific cell volume perturbations in initiation and progression of apoptosis in vascular smooth muscle cells should be examined further.


Subject(s)
Apoptosis , Cell Size , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cells, Cultured , Chromatin/metabolism , Colforsin/pharmacology , Culture Media, Serum-Free , DNA Cleavage , Myocytes, Smooth Muscle/drug effects , Osmotic Pressure , Ouabain/pharmacology , Rats , Single-Cell Analysis , Staurosporine/pharmacology , Time-Lapse Imaging
15.
Am J Physiol Cell Physiol ; 301(2): C403-12, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21562307

ABSTRACT

Purinergic receptors activate diverse signaling cascades and regulate the activity of cell volume-sensitive ion transporters. However, the effects of ATP and other agonists of P2 receptors on cell volume dynamics are only scarcely studied. In the present work, we used the recently developed dual-image surface reconstruction technique to explore the influence of purinergic agonists on cell volume in the C11-Madin-Darby canine kidney cell line resembling intercalated cells from kidney collecting ducts. Unexpectedly, we found that ATP and UTP triggered very robust (55-60%) cell shrinkage that lasted up to 2 h after agonist washout. Purinergic regulation of cell volume required increases in intracellular Ca(2+) and could be partially mimicked by the Ca(2+)-ionophore ionomycin or activation of protein kinase C by 4ß-phorbol 12-myristate 13-acetate. Cell shrinkage was accompanied by strong reductions in intracellular K(+) and Cl(-) content measured using steady-state (86)Rb(+) and (36)Cl(-) distribution. Both shrinkage and ion efflux in ATP-treated cells were prevented by the anion channel blocker 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB) and by the BK(Ca) channel inhibitors charybdotoxin, iberiotoxin, and paxilline. To evaluate the significance of cell-volume changes in purinergic signaling, we measured the impact of ATP on the expression of the immediate-early gene c-Fos. Thirty-minute treatment with ATP increased c-Fos immunoreactivity by approximately fivefold, an effect that was strongly inhibited by charybdotoxin and completely prevented by NPPB. Overall, our findings suggest that ATP-induced cell-volume changes are partially responsible for the physiological actions of purinergic agonists.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Size , Epithelial Cells/metabolism , Kidney/metabolism , Purinergic P2Y Receptor Agonists/metabolism , Receptors, Purinergic P2Y/metabolism , Uridine Triphosphate/metabolism , Animals , Calcium Signaling , Cell Line , Chlorides/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dogs , Enzyme Activation , Enzyme Activators/pharmacology , Epithelial Cells/drug effects , Ionophores/pharmacology , Kidney/cytology , Kidney/drug effects , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Time Factors
16.
J Membr Biol ; 241(3): 145-54, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21584679

ABSTRACT

This study examined the role of cell volume modulation in plasma membrane rupture and death documented in ouabain-treated renal epithelial cells. Long-term exposure to ouabain caused massive death of C11-MDCK (Madin-Darby canine kidney) epithelial cells, documented by their detachment, chromatin cleavage and complete loss of lactate dehydrogenase (LDH), but did not affect the survival of vascular smooth muscle cells (VSMCs) from the rat aorta. Unlike the distinct impact on cell survival, 2-h exposure to ouabain led to sharp elevation of the [Na⁺](i)/[K⁺](i) ratio in both cell types. A similar increment of Na⁺(i) content was evoked by sustained inhibition of Na⁺,K⁺-ATPase in K⁺-free medium. However, in contrast to ouabain, C11-MDCK cells survived perfectly during 24-h exposure to K⁺-free medium. At 3 h, the volume of ouabain-treated C11-MDCK cells and VSMCs, measured by the recently developed dual-image surface reconstruction technique, was increased by 16 and 12%, respectively, whereas 5-10 min before the detachment of ouabain-treated C11-MDCK cells, their volume was augmented by ~30-40%. To examine the role of modest swelling in the plasma membrane rupture of ouabain-treated cells, we compared actions of hypotonic medium on volume and LDH release. We observed that LDH release from hypoosmotically swollen C11-MDCK cells was triggered when their volume was increased by approximately fivefold. Thus, our results showed that the rupture of plasma membranes in ouabain-treated C11-MDCK cells was not directly caused by cell volume modulation evoked by Na⁺,K⁺-ATPase inhibition and inversion of the [Na⁺](i)/[K⁺](i) ratio.


Subject(s)
Cell Membrane/metabolism , Cell Size/drug effects , Epithelial Cells/metabolism , Ion Transport/drug effects , Kidney/metabolism , Muscle, Smooth, Vascular/metabolism , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase , Animals , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Chromatin/metabolism , Dogs , Epithelial Cells/cytology , Kidney/cytology , L-Lactate Dehydrogenase/metabolism , Muscle, Smooth, Vascular/cytology , Osmosis/drug effects , Osmotic Pressure/drug effects , Potassium/metabolism , Rats , Rubidium/metabolism , Sodium Isotopes/metabolism , Sodium Radioisotopes/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism
17.
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.

18.
Cell Physiol Biochem ; 23(4-6): 407-14, 2009.
Article in English | MEDLINE | ID: mdl-19471108

ABSTRACT

In smooth muscles, inhibition of Na(+),K(+),2Cl(-) cotransport (NKCC) by bumetanide decreased intracellular Cl(-) content ([Cl(-)](i)) and suppressed the contractions triggered by diverse stimuli. This study examines whether or not bicarbonate, a regulator of several Cl(-) transporters, affects the impact of NKCC in excitation-contraction coupling. Addition of 25 mM NaHCO(3) attenuated the inhibitory action of bumetanide on mesenteric artery contractions evoked by 30 mM KCl and phenylephrine (PE) by 5 and 3-fold, respectively. In cultured vascular smooth muscle cells, NaHCO(3) almost completely abolished inhibitory actions of bumetanide on transient depolarization and [Ca(2+)](i) elevation triggered by PE. In bicarbonate-free medium, bumetanide decreased [Cl(-)](i) by approximately 15%; this effect was almost totally abrogated by NaHCO(3). The addition of NaHCO(3) resulted in 2-fold inhibition of NKCC activity and 3-fold attenuation of [Cl(-)](i). These data strongly suggest that extracellular HCO(3)(-) diminishes the NKCC-sensitive component of excitation-contraction coupling via suppression of this carrier.


Subject(s)
Bicarbonates/pharmacology , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Bumetanide/pharmacology , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Phenylephrine/pharmacology
19.
Purinergic Signal ; 5(3): 343-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19387869

ABSTRACT

This study examines the action of agonists and antagonists of P2 receptors on mouse mesenteric artery contractions and the possible involvement of these signaling pathways in myogenic tone (MT) evoked by elevated intraluminal pressure. Both ATP and its non-hydrolyzed analog alpha,beta-ATP triggered transient contractions that were sharply decreased in the presence of NF023, a potent antagonist of P2X(1) receptors. In contrast, UTP and UDP elicited sustained contractions which were suppressed by MRS2567, a selective antagonist of P2Y(6) receptors. Inhibition of Na(+), K(+), 2Cl(-) cotransport (NKCC) with bumetanide led to attenuation of contractions in UTP- but not ATP-treated arteries. Both UTP-induced contractions and MT were suppressed by MRS2567 and bumetanide but were insensitive to NF023. These data implicate a P2Y(6)-mediated, NKCC-dependent mechanism in MT of mesenteric arteries. The action of heightened intraluminal pressure on UTP release from mesenteric arteries and its role in the triggering of P2Y(6)-mediated signaling should be examined further.

20.
Biochem Biophys Res Commun ; 379(4): 1080-3, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19150334

ABSTRACT

Bumetanide and other high-ceiling diuretics (HCD) attenuate myogenic tone and contractions of vascular smooth muscle cells (VSMC) triggered by diverse stimuli. HCD outcome may be mediated by their interaction with NKCC1, the only isoform of Na(+), K(+), 2Cl(-) cotransporter expressed in VSMC as well as with targets distinct from this carrier. To examine these hypotheses, we compared the effect of bumetanide on contractions of mesenteric arteries from wild-type and NKCC1 knockout mice. In mesenteric arteries from wild-type controls, 100 microM bumetanide evoked a decrease of up to 4-fold in myogenic tone and contractions triggered by modest [K(+)](o)-induced depolarization, phenylephrine and UTP. These actions of bumetanide were preserved after inhibition of nitric oxide synthase with NG-nitro-l-arginine methyl ester, but were absent in mesenteric arteries from NKCC1(-/-) mice. The data show that bumetanide inhibits VSMC contractile responses via its interaction with NKCC1 and independently of nitric oxide production by endothelial cells.


Subject(s)
Mesenteric Arteries/drug effects , Muscle, Smooth, Vascular/drug effects , Sodium Potassium Chloride Symporter Inhibitors , Vascular Resistance/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects , Animals , Bumetanide/pharmacology , Diuretics/pharmacology , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Muscle Tonus/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Nitric Oxide/metabolism , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Sodium-Potassium-Chloride Symporters/physiology , Solute Carrier Family 12, Member 2
SELECTION OF CITATIONS
SEARCH DETAIL
...