Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 318(3): C581-C597, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31913698

ABSTRACT

The histone deacetylase inhibitor trichostatin A (TSA) reduces cell viability in cisplatin-sensitive (A2780WT) and cisplatin-resistant (A2780RES) human ovarian cancer cells due to progression of apoptosis (increased caspase-9 activity), autophagy (increased LC3-II expression), and cell cycle arrest (increased p21 expression). The TSA-mediated effect on p21 and caspase-9 is mainly p53 independent. Cisplatin increases DNA-damage (histone H2AX phosphorylation) in A2780WT cells, whereas cisplatin, due to reduced uptake [inductively coupled-plasma-mass spectrometry (Pt) analysis], has no DNA-damaging effect in A2780RES cells. TSA has no effect on cisplatin accumulation or cisplatin-induced DNA-damage in A2780WT/A2780RES cells. Tracer technique indicates that TSA inhibits the volume-sensitive organic anion channel (VSOAC) in A2780WT/A2780RES cells and that the activity is restored by exogenous H2O2. As TSA reduces NOX4 mRNA accumulation and concomitantly increases catalase mRNA/protein accumulation, we suggest that TSA increases the antioxidative defense in A2780 cells. Inhibition of the kinase mTOR (rapamycin, palomid, siRNA), which is normally associated with cell growth, reduces VSOAC activity synergistically to TSA. However, as TSA increases mTOR activity (phosphorylation of 4EBP1, S6 kinase, S6, ULK1, SGK1), the effect of TSA on VSOAC activity does not reflect the shift in mTOR signaling. Upregulation of the protein expression and activity of the taurine transporter (TauT) is a phenotypic characteristic of A2780RES cells. However, TSA reduces TauT protein expression in A2780RES cells and activity to values seen in A2780WT cells. It is suggested that therapeutic benefits of TSA in A2780 do not imply facilitation of cisplatin uptake but more likely a synergistic activation of apoptosis/autophagy and reduced TauT activity.


Subject(s)
Cell Size/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Ovarian Neoplasms/metabolism , Taurine/metabolism , A549 Cells , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Female , Humans , Ovarian Neoplasms/pathology
2.
J Pharm Biomed Anal ; 165: 82-89, 2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30508755

ABSTRACT

The feasibility of quantitatively tracking platinum, derived from platinum-based compounds, during subcellular fractionation was studied. Cisplatin-exposed murine Ehrlich Lettré Ascites cells were fractionated into cytosolic and crude nuclear fractions. The latter was subsequently purified. Every residue and fraction produced during the fractionation procedure were collected and the platinum content determined by inductively coupled plasma mass spectrometry. Western blotting verified that the nuclear and cytosolic fractions were pure. It was found that 18% of platinum taken up by the cells was located in the nuclear fraction while 66% was located in the cytosolic fraction. Accumulated uncertainty originating from invariable sample characteristics and giving fraction purity priority had a negative effect on platinum recovery. Thus, overall 81% (n = 3, RSD = 3.4%) of the platinum taken up by the cells was recovered in the residues and final fractions. In conclusion, a reliable intracellular localization and quantitation of platinum following administration of Cisplatin can be determined by application of the method.


Subject(s)
Antineoplastic Agents/metabolism , Carcinoma, Ehrlich Tumor/metabolism , Cisplatin/metabolism , Mass Spectrometry/methods , Animals , Blotting, Western , Cell Nucleus/metabolism , Cytosol/metabolism , Mice , Subcellular Fractions/metabolism , Tumor Cells, Cultured
3.
Int J Mol Sci ; 19(8)2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30071606

ABSTRACT

Cisplatin, carboplatin, and oxaliplatin are Pt-based drugs used in the chemotherapeutic eradication of cancer cells. Although most cancer patient cells initially respond well to the treatment, the clinical effectiveness declines over time as the cancer cells develop resistance to the drugs. The Pt-based drugs are accumulated via membrane-bound transporters, translocated to the nucleus, where they trigger various intracellular cell death programs through DNA interaction. Here we illustrate how resistance to Pt-based drugs, acquired through limitation in the activity/subcellular localization of canonical drug transporters, might be circumvented by the facilitated uptake of Pt-based drug complexes via nanocarriers/endocytosis or lipophilic drugs by diffusion.


Subject(s)
Carboplatin/pharmacokinetics , Cell Membrane , Cell Nucleus , Cisplatin/pharmacokinetics , Neoplasms , Organoplatinum Compounds/pharmacokinetics , Animals , Carboplatin/therapeutic use , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cisplatin/therapeutic use , DNA, Neoplasm/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Organoplatinum Compounds/therapeutic use , Oxaliplatin
4.
J Inorg Biochem ; 160: 287-95, 2016 07.
Article in English | MEDLINE | ID: mdl-27112899

ABSTRACT

Acquired resistance to chemotherapeutic drugs in cancer cells can reflect an ability to limit cellular drug availability, to repair drug induced DNA damage, and to limit initiation/progression of cell death (apoptosis). The leucine-rich-repeat-containing 8A (LRRC8A) protein is an essential component of volume sensitive channels for organic osmolytes (VSOAC) and volume regulated anion channels (VRAC), which are activated during the apoptotic process. Here we illustrate that cisplatin resistance in human ovarian cancer cells (A2780) correlates with a reduced expression of LRRC8A and copper transporter receptor 1 (CTR1), as well as a concomitant increased expression of copper-transporting P-type ATPases (ATP7A/ATP7B). We also find that cisplatin (Pt) accumulation correlates with LRRC8A protein expression and channel activity, i.e., the cellular Pt content is high when VSOAC is activated by depolarization of the plasma membrane or hypoosmotic cell swelling, and reduced when channel activity/LRRC8A expression is reduced by genetically silencing/pharmacological inhibition, or the cells have acquired a resistant phenotype with low LRRC8A protein expression. It is suggested that reduced LRRC8A expression in cisplatin-resistant A2780 cells ensures cell survival through limitation in cisplatin accumulation and a concomitant reduction in osmolytes loss via VSOAC/VRAC and hence instigation of the apoptotic process.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Survival/genetics , Copper Transporter 1 , Copper-Transporting ATPases , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
5.
Am J Physiol Cell Physiol ; 310(11): C857-73, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26984736

ABSTRACT

The leucine-rich repeat containing 8A (LRRC8A) protein is an essential component of the volume-sensitive organic anion channel (VSOAC), and using pharmacological anion channel inhibitors (NS3728, DIDS) and LRRC8A siRNA we have investigated its role in development of Cisplatin resistance in human ovarian (A2780) and alveolar (A549) carcinoma cells. In Cisplatin-sensitive cells Cisplatin treatment increases p53-protein level as well as downstream signaling, e.g., expression of p21(Waf1/Cip1), Bax, Noxa, MDM2, and activation of Caspase-9/-3. In contrast, Cisplatin-resistant cells do not enter apoptosis, i.e., their p53 and downstream signaling are reduced and caspase activity unaltered following Cisplatin exposure. Reduced LRRC8A expression and VSOAC activity are previously shown to correlate with Cisplatin resistance, and here we demonstrate that pharmacological inhibition and transient knockdown of LRRC8A reduce the protein level of p53, MDM2, and p21(Waf1/Cip1) as well as Caspase-9/-3 activation in Cisplatin-sensitive cells. Cisplatin resistance is accompanied by reduction in total LRRC8A expression (A2780) or LRRC8A expression in the plasma membrane (A549). Activation of Caspase-3 dependent apoptosis by TNFα-exposure or hyperosmotic cell shrinkage is almost unaffected by pharmacological anion channel inhibition. Our data indicate 1) that expression/activity of LRRC8A is essential for Cisplatin-induced increase in p53 protein level and its downstream signaling, i.e., Caspase-9/-3 activation, expression of p21(Waf1/Cip1) and MDM2; and 2) that downregulation of LRRC8A-dependent osmolyte transporters contributes to acquirement of Cisplatin resistance in ovarian and lung carcinoma cells. Activation of LRRC8A-containing channels is upstream to apoptotic volume decrease as hypertonic cell shrinkage induces apoptosis independent of the presence of LRRC8A.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Antineoplastic Agents/pharmacology , Caspase 3/metabolism , Caspase 9/metabolism , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Lung Neoplasms/drug therapy , Membrane Proteins/metabolism , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/enzymology , Adenocarcinoma, Bronchiolo-Alveolar/genetics , Adenocarcinoma, Bronchiolo-Alveolar/pathology , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/metabolism , Apoptosis/drug effects , Caspase 3/genetics , Caspase 9/genetics , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/metabolism , Cell Size , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Proteins/genetics , Membrane Transport Modulators/pharmacology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics
6.
Metallomics ; 8(3): 298-304, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26739575

ABSTRACT

Cellular uptake of vitamin B12-cisplatin conjugates was estimated via detection of their metal constituents (Co, Pt, and Re) by inductively coupled plasma mass spectrometry (ICP-MS). Vitamin B12 (cyano-cob(iii)alamin) and aquo-cob(iii)alamin [Cbl-OH2](+), which differ in the ß-axial ligands (CN(-) and H2O, respectively), were included as control samples. The results indicated that B12 derivatives delivered cisplatin to both cellular cytosol and nuclei with an efficiency of one third compared to the uptake of free cisplatin cis-[Pt(II)Cl2(NH3)2]. In addition, uptake of charged B12 derivatives including [Cbl-OH2](+), [{Co}-CN-{cis-PtCl(NH3)2}](+), [{Re}-{Co}-CN-{cis-PtCl(NH3)2}](+), and [{Co}-CN-{trans-Pt(Cyt)(NH3)2}](2+) (Cyt = cytarabin) was high compared to neutral B12, which implied the existence of an additional internalization pathway for charged B12 vitamin analogs. The affinities of the charged B12 derivatives to the B12 transporters HC, IF and TC were similar to that of native vitamin B12.


Subject(s)
Endocytosis , Metals/metabolism , Vitamin B 12/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/metabolism , Endocytosis/drug effects , Humans , K562 Cells , Protein Transport/drug effects , Proton Magnetic Resonance Spectroscopy , Vitamin B 12/chemical synthesis , Vitamin B 12/chemistry , Vitamin B 12/pharmacology
7.
Phytother Res ; 30(1): 97-104, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26549524

ABSTRACT

We have tested the effect of protolichesterinic acid (PA) on the activity of the volume-sensitive release pathway for the organic osmolyte taurine (VSOAC) and the expression of the leucine-rich-repeat-channel 8A (LRRC8A) protein, which constitutes an essential VSOAC component. Exposing human lung cancer cells (A549) to PA (20 µg/mL, 24 h) reduces LRRC8A protein expression by 25% and taurine release following osmotic cell swelling (320 → 200 mOsm) by 60%. C75 (20 µg/mL, 24 h), a γ-lactone with a C8 carbon fatty acid chain, reduces VSOAC activity by 30%, i.e. less than PA. Stearic acid (20 µg/mL, 24 h) has no effect on VSOAC. Hence, length of PA's fatty acid chain adds to γ-lactone's inhibitory action. 5-Lipoxygenase (5-LO) activity is essential for swelling-induced activation of VSOAC. PA has no effect on cellular concentration of leukotrienes (5-HETE/LTB4 ) under hypotonic conditions, excluding that PA mediated inhibition of VSOAC involves 5-LO inhibition. A549 cells exposed to the chemotherapeutic drug cisplatin (10 µM, 24 h) reveal signs of apoptosis, i.e. 25% reduction in cell viability as well as 1.3-, 1.5- and 3.3-fold increase in the expression of LRRC8A, Bax (regulator of apoptosis) and p21 (regulator of cell cycle progression), respectively. PA reduces cell viability by 30% but has no effect on p21/Bax expression. This excludes PA as a pro-apoptotic drug in A549 cells.


Subject(s)
4-Butyrolactone/analogs & derivatives , Lichens/chemistry , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Parmeliaceae/chemistry , Taurine/metabolism , 4-Butyrolactone/pharmacology , Apoptosis/drug effects , Arachidonate 5-Lipoxygenase/metabolism , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Hydroxyeicosatetraenoic Acids/metabolism , Lung Neoplasms/pathology , bcl-2-Associated X Protein/metabolism
8.
Physiol Rep ; 3(6)2015 Jun.
Article in English | MEDLINE | ID: mdl-26056062

ABSTRACT

The aim of this project was to analyze the regulation of p53 expression in NIH3T3 fibroblasts under the influence of increasing hyperosmotic stress. Expression of p53 showed a biphasic response pattern in NIH3T3 cells under increasing osmotic stress (337 mOsm to 737 mOsm) with a maximum at 587 mOsm. Under isotonic conditions p53 expression increased after addition of the proteasome inhibitor MG132 indicating that cellular p53 levels in unperturbed cells is kept low by proteasomal degradation. However, under hypertonic conditions p53 synthesis as well as p53 degradation were significantly reduced and it is demonstrated that the increase in p53 expression observed when tonicity is increased from 337 to 587 mOsm reflects that degradation is more inhibited than synthesis, whereas the decrease in p53 expression at higher tonicities reflects that synthesis is more inhibited than degradation. The activity of the p53 regulating proteins p38 MAP kinase and the ubiquitin ligase MDM2 were studied as a function of increasing osmolarity. MDM2 protein expression was unchanged at all osmolarities, whereas MDM2 phosphorylation (Ser(166)) increased at osmolarities up to 537 mOsm and remained constant at higher osmolarities. Phosphorylation of p38 increased at osmolarities up to 687 mOsm which correlated with an increased phosphorylation of p53 (Ser(15)) and the decreased p53 degradation. Caspase-3 activity increased gradually with hypertonicity and at 737 mOsm both Caspase-3 activity and annexin V binding are high even though p53 expression and activity are low, indicating that initiation of apoptosis under severe hypertonic conditions is not strictly controlled by p53.

9.
Cell Physiol Biochem ; 36(1): 111-32, 2015.
Article in English | MEDLINE | ID: mdl-25925201

ABSTRACT

BACKGROUND/AIMS: Altered expression of the integrin family of cell adhesion receptors has been associated with initiation, progression, and metastasis of solid tumors as well as in the development of chemoresistance. Here, we investigated the role of integrins, in particular integrin ß1, in cell volume regulation and drug-induced apoptosis in adherent and non-adherent Ehrlich ascites cell lines. METHODS: Adhesion phenotypes were verified by colorimetric cell-adhesion-assay. Quantitative real-time PCR and western blot were used to compare expression levels of integrin subunits. Small interfering RNA was used to silence integrin ß1 expression. Regulatory volume decrease (RVD) after cell swelling was studied with calcein-fluorescence-self-quenching and Coulter counter analysis. Taurine efflux was estimated with tracer technique. Caspase assay was used to determine apoptosis. RESULTS: We show that adherent cells have stronger fibronectin binding and a significantly increased expression of integrin α5, αv, and ß1 at mRNA and protein level, compared to non-adherent cells. Knockdown of integrin ß1 reduced RVD of the adherent but not of the non-adherent cells. Efflux of taurine was unaffected. In contrast to non-adherent, adherent cells exhibited chemoresistance to chemotherapeutic drugs (cisplatin and gemcitabine). However, knockdown of integrin ß1 promoted cisplatin-induced caspase activity in adherent cells. CONCLUSION: Our data identifies integrin ß1 as a part of the osmosensing machinery and regulator of cisplatin resistance in adherent Ehrlich cells.


Subject(s)
Carcinoma, Ehrlich Tumor/metabolism , Drug Resistance, Neoplasm , Integrin beta1/genetics , Integrin beta1/metabolism , Osmosis , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Ehrlich Tumor/genetics , Carcinoma, Ehrlich Tumor/pathology , Caspases/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Size/drug effects , Cisplatin/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Fibronectins/metabolism , Mice , Taurine/metabolism , Gemcitabine
10.
Metallomics ; 7(5): 885-95, 2015 May.
Article in English | MEDLINE | ID: mdl-25805368

ABSTRACT

In the present work a novel C,N-cyclometalated benzimidazole Ru(ii) arene complex (GY34) was characterized by applying an alternative, diverse approach considering both chemical and biological aspects. RP-HPLC-ICP-MS and RP-HPLC-ESI-MS analysis proved that GY34 in both RPMI-1640 cell medium and ammonium acetate buffer was transformed into several subspecies and the importance of evaluating and controlling analyte stability throughout experiments was demonstrated. Applying a novel cell fractionation protocol GY34 was found to target cell nuclei and mitochondria in Ehrlich Lettré Ascites (ELA) cells, with the intracellular distribution depending on GY34 concentration in the cell medium during incubation. In ELA cells 96 ± 0.2% of cytosolic GY34 was bound to high-molecular species. Furthermore, using the tracer technique GY34 was found to reduce uptake and increase release of the organic osmolyte taurine in ELA cells, with innate resistance to Cisplatin and in A2780 human ovarian cancer cells, with acquired resistance to Cisplatin. Importantly, FACS analysis revealed that GY34 induced apoptosis in ELA cells. The present data suggest the potential of GY34 in overcoming Cisplatin resistance. The methodology applied can be used as a general protocol and an additional tool in the initial evaluation of novel metal-based drugs.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Cell Line, Tumor , Female , Homeostasis/drug effects , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Ruthenium/pharmacokinetics , Taurine/metabolism
11.
Am J Physiol Cell Physiol ; 307(12): C1071-80, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25252947

ABSTRACT

Cisplatin resistance is a major challenge in the treatment of cancer and develops through reduced drug accumulation and an increased ability to avoid drug-induced cell damage, cell shrinkage, and hence initiation of apoptosis. Uptake and release of the semiessential amino acid taurine contribute to cell volume homeostasis, and taurine has been reported to have antiapoptotic effects. Here we find that volume-sensitive taurine release in cisplatin-sensitive [wild-type (WT)] human ovarian cancer A2780 cells is reduced in the presence of the phospholipase A2 inhibitor bromenol lactone, the 5-lipoxygenase (5-LO) inhibitor ETH 615-139, and the cysteine leukotriene receptor 1 (CysLT1) antagonist zafirlukast and impaired by the anion channel blocker DIDS (4,4'-diisothiocyanatostilbene-2,2'-disulfonate). Comparing WT and cisplatin-resistant (RES) A2780 cells we also find that evasion of cisplatin-induced cell death in RES A2780 cells correlates with an increased accumulation of taurine, due to an increased taurine uptake and a concomitant impairment of the volume-sensitive taurine release pathway, as well an inability to reduce cell volume after osmotic cell swelling. Downregulation of volume-sensitive taurine release in RES A2780 cells correlates with reduced expression of the leucine-rich repeat-containing protein 8A (LRRC8A). Furthermore, acute (18 h) exposure to cisplatin (5-10 µM) increases taurine release and LRRC8A expression in WT A2780 cells whereas cisplatin has no effect on LRRC8A expression in RES A2780 cells. It is suggested that shift in LRRC8A activity can be used as biomarker for apoptotic progress and acquirement of drug resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Size/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Taurine/metabolism , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation , Female , Homeostasis , Humans , Leukotriene Antagonists/pharmacology , Lipoxygenase Inhibitors/pharmacology , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Osmotic Pressure , Ovarian Neoplasms/pathology , Phospholipase A2 Inhibitors/pharmacology , Time Factors
12.
IUBMB Life ; 66(4): 257-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24771413

ABSTRACT

The review describes molecular and functional properties of the volume regulated anion channel and Ca(2+)-dependent Cl(-) channels belonging to the anoctamin family with emphasis on physiological importance of these channels in regulation of cell volume, cell migration, cell proliferation, and programmed cell death. Finally, we discuss the role of Cl(-) channels in various diseases.


Subject(s)
Anions/metabolism , Calcium/metabolism , Cell Physiological Phenomena , Chloride Channels/metabolism , Animals , Humans
13.
Am J Physiol Cell Physiol ; 306(11): C1028-40, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24696147

ABSTRACT

Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that modulates translation in response to growth factors and alterations in nutrient availability following hypoxia and DNA damage. Here we demonstrate that mTOR activity in Ehrlich Lettré ascites (ELA) cells is transiently increased within minutes following osmotic cell swelling and that inhibition of phosphatidylinositol-3-phosphatase (PTEN) counteracts the upstream phosphatidylinositol kinase and potentiates mTOR activity. PTEN inhibition concomitantly potentiates swelling-induced taurine release via the volume-sensitive transporter for organic osmolytes and anion channels (VSOAC) and enhances swelling-induced inhibition of taurine uptake via the taurine-specific transporter (TauT). Chronic osmotic stress, i.e., exposure to hypotonic or hypertonic media for 24 h, reduces and increases mTOR activity in ELA cells, respectively. Using rapamycin, we demonstrate that mTOR inhibition is accompanied by reduction in TauT activity and increase in VSOAC activity in cells expressing high (NIH3T3 fibroblasts) or low (ELA) amounts of mTOR protein. The effect of mTOR inhibition on TauT activity reflects reduced TauT mRNA, TauT protein abundance, and an overall reduction in protein synthesis, whereas the effect on VSOAC is mimicked by catalase inhibition and correlates with reduced catalase mRNA abundance. Hence, mTOR activity favors loss of taurine following hypoosmotic cell swelling, i.e., release via VSOAC and uptake via TauT during acute hypotonic exposure is potentiated and reduced, respectively, by phosphorylation involving mTOR and/or the kinases upstream to mTOR. Decrease in TauT activity during chronic hypotonic exposure, on the other hand, involves reduction in expression/activity of TauT and enzymes in antioxidative defense.


Subject(s)
Fibroblasts/metabolism , Osmotic Pressure/physiology , TOR Serine-Threonine Kinases/metabolism , Taurine/metabolism , Animals , Biological Transport/physiology , Mice , NIH 3T3 Cells , Osmosis/physiology
14.
Am J Physiol Cell Physiol ; 305(1): C48-60, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23485709

ABSTRACT

Pathophysiological conditions challenge cell volume homeostasis and perturb cell volume regulatory mechanisms leading to alterations of cell metabolism, active transepithelial transport, cell migration, and death. We report that inhibition of the 5-lipoxygenase (5-LO) with AA861 or ETH 615-139, the cysteinyl leukotriene 1 receptor (CysLT1) with the antiasthmatic drug Zafirlukast, or the volume-sensitive organic anion channel (VSOAC) with DIDS blocks the release of organic osmolytes (taurine, meAIB) and the concomitant cell volume restoration following hypoosmotic swelling of human type II-like lung epithelial cells (A549). Reactive oxygen species (ROS) are produced in A549 cells upon hypotonic cell swelling by a diphenylene iodonium-sensitive NADPH oxidase. The swelling-induced taurine release is suppressed by ROS scavenging (butylated hydroxytoluene, N-acetyl cysteine) and potentiated by H2O2. Ca²âº mobilization with ionomycin or ATP stimulates the swelling-induced taurine release whereas calmodulin inhibition (W7) inhibits the release. Chelation of the extracellular Ca²âº (EGTA) had no effect on swelling-induced taurine release but prevented ATP-induced stimulation. H2O2, ATP, and ionomycin were unable to stimulate the taurine release in the presence of AA861 or Zafirlukast, placing 5-LO and CysLT1 as essential elements in the swelling-induced activation of VSOAC with ROS and Ca²âº as potent modulators. Inhibition of tyrosine kinases (genistein, cucurbitacin) reduces volume-sensitive taurine release, adding tyrosine kinases (Janus kinase) as regulators of VSOAC activity. Caspase-3 activity during hypoxia is unaffected by inhibition of 5-LO/CysLT1 but reduced when swelling-induced taurine loss via VSOAC is prevented by DIDS excess extracellular taurine, indicating a beneficial role of taurine under hypoxia.


Subject(s)
Amino Acids/metabolism , Arachidonate 5-Lipoxygenase/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Adenocarcinoma , Adenosine Triphosphate , Anti-Asthmatic Agents/pharmacology , Arachidonate 5-Lipoxygenase/genetics , Calcium , Cell Line, Tumor , Cell Size/drug effects , Cell Survival , Dose-Response Relationship, Drug , Electrolytes , Gene Expression Regulation, Enzymologic , Humans , Indoles , Leukotriene Antagonists/pharmacology , Lipoxygenase Inhibitors/pharmacology , Lung Neoplasms , Osmolar Concentration , Osmotic Pressure/physiology , Phenylcarbamates , Reactive Oxygen Species/metabolism , Sulfonamides , Taurine/metabolism , Tosyl Compounds/pharmacology , Water/metabolism
15.
J Membr Biol ; 245(2): 77-87, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22383044

ABSTRACT

The present work was initiated to investigate regulation of the taurine transporter TauT by reactive oxygen species (ROS) and the tonicity-responsive enhancer binding protein (TonEBP) in NIH3T3 mouse fibroblasts during acute and long-term (4 h) exposure to low-sodium/hypo-osmotic stress. Taurine influx is reduced following reduction in osmolarity, keeping the extracellular Na(+) concentration constant. TonEBP activity is unaltered, whereas TauT transcription as well as TauT activity are significantly reduced under hypo-osmotic conditions. In contrast, TonEBP activity and TauT transcription are significantly increased following hyperosmotic exposure. Swelling-induced ROS production in NIH3T3 fibroblasts is generated by NOX4 and by increasing total ROS, by either exogenous application of H(2)O(2) or overexpressing NOX4, we demonstrate that TonEBP activity and taurine influx are regulated negatively by ROS under hypo-osmotic, low-sodium conditions, whereas the TauT mRNA level is unaffected. Acute exposure to ROS reduces taurine uptake as a result of modulated TauT transport kinetics. Thus, swelling-induced ROS production could account for the reduced taurine uptake under low-sodium/hypo-osmotic conditions by direct modulation of TauT.


Subject(s)
Fibroblasts/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Stress, Physiological/physiology , Animals , Cells, Cultured , Down-Regulation , Hydrogen Peroxide/metabolism , Hypertonic Solutions , Hypotonic Solutions , Kinetics , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Mice , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NIH 3T3 Cells , Osmosis , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Sodium/metabolism , Taurine/genetics , Taurine/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Water-Electrolyte Balance
16.
Cell Physiol Biochem ; 28(6): 1099-110, 2011.
Article in English | MEDLINE | ID: mdl-22178999

ABSTRACT

Maintaining cell volume is critical for cellular function yet shift in cell volume is a prerequisite for mitosis and apoptosis. The ubiquitously and evolutionary conserved serine/threonine kinase CK2 promotes cell survival and suppresses apoptosis. The present review describes how mammalian cells regulate the cellular content of the major cellular organic osmolyte, taurine with emphasis on CK2 mediated regulation of active taurine uptake and volume-sensitive taurine release. Furthermore, we discuss how CK2-mediated regulation of taurine homeostasis is potentially involved in cellular functions such as proliferation and survival.


Subject(s)
Casein Kinase II/metabolism , Taurine/metabolism , Animals , Cell Size , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/physiology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism
17.
Amino Acids ; 40(4): 1091-106, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20827495

ABSTRACT

Increased expression of the ubiquitous serine/threonine protein kinase CK2 has been associated with increased proliferative capacity and increased resistance towards apoptosis. Taurine is the primary organic osmolyte involved in cell volume control in mammalian cells, and shift in cell volume is a critical step in cell proliferation, differentiation and induction of apoptosis. In the present study, we use mouse NIH3T3 fibroblasts and Ehrlich Lettré ascites tumour cells with different CK2 expression levels. Taurine uptake via the Na(+) dependent transporter TauT and taurine release are increased and reduced, respectively, following pharmacological CK2 inhibition. The effect of CK2 inhibition on TauT involves modulation of transport kinetics, whereas the effect on the taurine release pathway involves reduction in the open-probability of the efflux pathway. Stimulation of PLA(2) activity, exposure to exogenous reactive oxygen species as well as inhibition of protein tyrosine phosphotases (PTP) potentiate the swelling-induced taurine loss. Inhibition of PI3K and PTEN reduces and potentiates swelling-induced taurine release, respectively. Inhibition of CK2 has no effect on PLA(2) activity and ROS production by NADPH oxidase, whereas it lifts the effect of PTEN and PTP inhibition. It is suggested that CK2 regulates the taurine release downstream to known swelling-induced signal transducers including PLA(2), NADPH oxidase and PI3K.


Subject(s)
Casein Kinase II/metabolism , Fibroblasts/metabolism , Protein Kinase Inhibitors/pharmacology , Taurine/metabolism , Animals , Apoptosis/drug effects , Biological Transport/drug effects , Carcinoma, Ehrlich Tumor/metabolism , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Cell Size/drug effects , Gene Expression , Homeostasis , Kinetics , Mice , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NIH 3T3 Cells , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phospholipases A2/genetics , Phospholipases A2/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Water-Electrolyte Balance/drug effects
18.
Metallomics ; 2(12): 811-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21085722

ABSTRACT

The accumulation and cytotoxicity of a 10 µmol L⁻¹ equimolar human serum albumin-cisplatin adduct (HSA-Pt) was investigated in suspension Ehrlich Ascites Tumor Cells (EATC) and adherent Ehrlich Lettré Ascites Cells (Lettré). HSA-Pt did not induce apoptosis nor was it taken up by the cells to any significant amount within 24 h incubation. The accumulation and cytotoxicity of HSA-Pt was compared to 10 µmol L⁻¹ cisplatin for which a larger accumulation and cytotoxicity were observed in EATC compared to Lettré. The experiment was performed with cell medium exchange every fourth hour as HSA-Pt and cisplatin were not stable in RPMI-1640 with 10% serum. The stability was determined using size exclusion chromatography-inductively coupled plasma-mass spectrometry (SEC-ICP-MS) and after 4 h new platinum peaks were observed. These findings indicate that before conducting cell experiments, the stability of the compound in the cell medium should be investigated especially when long exposure times are applied. Furthermore, HSA-Pt was found to be stable in Hanks Balanced Saline Solution (HBSS) and in Phosphate Buffered Saline (PBS) at pH 5.3, 6.1 and 7.4. Thus, the shift in pH when HSA-cisplatin passes from blood (pH 7.4) to tumor tissue (pH 5-6) is not capable of releasing cisplatin from HSA.


Subject(s)
Cisplatin/toxicity , Cytotoxins/toxicity , Serum Albumin/toxicity , Cell Line, Tumor , Cisplatin/chemistry , Cisplatin/metabolism , Cytotoxins/chemistry , Cytotoxins/metabolism , Humans , Metalloproteins/metabolism , Metalloproteins/toxicity , Platinum/chemistry , Platinum/metabolism , Platinum/toxicity , Serum Albumin/chemistry , Serum Albumin/metabolism
19.
Am J Physiol Cell Physiol ; 299(4): C844-53, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20631251

ABSTRACT

The swelling-activated K(+) currents (I(K,vol)) in Ehrlich ascites tumor cells (EATC) has been reported to be through the two-pore domain (K(2p)), TWIK-related acid-sensitive K(+) channel 2 (TASK-2). The regulatory volume decrease (RVD), following hypotonic exposure in EATC, is rate limited by I(K,vol) indicating that inhibition of RVD reflects inhibition of TASK-2. We find that in EATC the tyrosine kinase inhibitor genistein inhibits RVD by 90%, and that the tyrosine phosphatase inhibitor monoperoxo(picolinato)-oxo-vanadate(V) [mpV(pic)] shifted the volume set point for inactivation of the channel to a lower cell volume. Swelling-activated K(+) efflux was impaired by genistein and the Src kinase family inhibitor 4-amino-5-(4-chloro-phenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2) and enhanced by the tyrosine phosphatase inhibitor mpV(pic). With the use of the TASK-2 inhibitor clofilium, it is demonstrated that mpV(pic) increased the volume-sensitive part of the K(+) efflux 1.3 times. To exclude K(+) efflux via a KCl cotransporter, cellular Cl(-) was substituted with NO(3)(-). Also under these conditions K(+) efflux was completely blocked by genistein. Thus tyrosine kinases seem to be involved in the activation of the volume-sensitive K(+) channel, whereas tyrosine phosphatases appears to be involved in inactivation of the channel. Overexpressing TASK-2 in human embryonic kidney (HEK)-293 cells increased the RVD rate and reduced the volume set point. TASK-2 has tyrosine sites, and precipitation of TASK-2 together with Western blotting and antibodies against phosphotyrosines revealed a cell swelling-induced, time-dependent tyrosine phosphorylation of the channel. Even though we found an inhibiting effect of PP2 on RVD, neither Src nor the focal adhesion kinase (FAK) seem to be involved. Inhibitors of the epidermal growth factor receptor tyrosine kinases had no effect on RVD, whereas the Janus kinase (JAK) inhibitor cucurbitacin inhibited the RVD by 40%. It is suggested that the cytokine receptor-coupled JAK/STAT pathway is upstream of the swelling-induced phosphorylation and activation of TASK-2 in EATC.


Subject(s)
Cell Size , Potassium Channels, Tandem Pore Domain/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Animals , Cell Line , Enzyme Inhibitors/metabolism , Female , Genistein/metabolism , Humans , Hypotonic Solutions , Janus Kinases/metabolism , Mice , Patch-Clamp Techniques , Phosphorylation , Potassium/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , STAT Transcription Factors/metabolism
20.
Amino Acids ; 39(5): 1521-36, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20499258

ABSTRACT

In mammalian cells, the organic osmolyte taurine is accumulated by the Na-dependent taurine transporter TauT and released though the volume- and DIDS-sensitive organic anion channel. Incubating Ehrlich Lettré tumor cells with methyl-ß-cyclodextrin (5 mM, 1 h) reduces the total cholesterol pool to 60±5% of the control value. Electron spin resonance data indicate a concomitant disruption of cholesterol-rich micro-domains. Active taurine uptake, cellular taurine content, and cell volume are reduced by 50, 20 and 20% compared to control values, respectively, whereas the passive taurine release is increased 4.5-fold under isotonic conditions following cholesterol depletion. However, taurine release under isotonic conditions is insensitive to DIDS and inhibitors of the volume-regulated anion channel. Uptake and release of meAIB are similarly affected following cholesterol depletion. Kinetic analysis reveals that cholesterol depletion increases TauT's affinity toward taurine but reduces its maximal transport capacity. Cholesterol depletion has no impact on TauT regulation by protein kinases A and C. Phospholipase A2 activity, which is required for the activation of volume-sensitive organic anion channel (VSOAC), is increased under isotonic and hypotonic conditions following cholesterol depletion, whereas taurine release under hypotonic conditions is reduced following cholesterol depletion. Hence, acute cholesterol depletion of Ehrlich Lettré cells leads to reduced TauT and VSOAC activities and at the same time increases the release of organic osmolytes via a leak pathway different from the volume-sensitive pathways for amino acids and anions.


Subject(s)
Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Cholesterol/deficiency , Cholesterol/metabolism , Taurine/metabolism , Animals , Ion Channels/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Mice , Osmosis , Phospholipases A2/metabolism , Tumor Cells, Cultured , beta-Cyclodextrins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...