Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Eur Respir J ; 39(2): 446-57, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21719484

ABSTRACT

Alveolar type II pneumocytes (ATII cells) are considered putative alveolar stem cells. Since no treatment is available to repair damaged epithelium and prevent lung fibrosis, novel approaches to induce regeneration of injured alveolar epithelium are desired. The objective of this study was to assess both the capacity of human embryonic stem cells (HUES-3) to differentiate in vitro into ATII cells and the ability of committed HUES-3 cells (HUES-3-ATII cells) to recover in vivo a pulmonary fibrosis model obtained by silica-induced damage. In vitro differentiated HUES-3-ATII cells displayed an alveolar phenotype characterised by multi-lamellar body and tight junction formation, by the expression of specific markers such as surfactant protein (SP)-B, SP-C and zonula occludens (ZO)-1 and the activity of cystic fibrosis transmembrane conductance regulator-mediated chloride ion transport. After transplantation of HUES-3-ATII cells into silica-damaged mice, histological and biomolecular analyses revealed a significant reduction of inflammation and fibrosis markers along with lung function improvement, weight recovery and increased survival. The persistence of human SP-C, human nuclear antigen and human DNA in the engrafted lungs indicates that differentiated cells remained engrafted up to 10 weeks. In conclusion, cell therapy using HUES-3 cells may be considered a promising approach to lung injury repair.


Subject(s)
Embryonic Stem Cells/transplantation , Pulmonary Fibrosis/therapy , Silicon Dioxide/toxicity , Silicosis/therapy , Stem Cell Transplantation/methods , Animals , Cell Differentiation/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Feeder Cells/cytology , Female , Fibroblasts/cytology , Humans , Mice , Mice, Nude , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Pulmonary Surfactant-Associated Protein C/metabolism , Silicosis/pathology , Treatment Outcome
2.
Biochem Biophys Res Commun ; 347(2): 452-9, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16824484

ABSTRACT

It has been shown that when CFTR and NHE3 are co-expressed on the apical membrane of the A6-NHE3 cell monolayers, the two transporters interact via a shared regulatory complex composed of NHERF2, ezrin, and PKA. We observe here that co-expression of NHE3 reduced both PKA-dependent apical CFTR expression and its activation once in place by approximately 50%. To analyze the role of NHERF2 in this process, we transfected NHE3 expressing and non-expressing A6 monolayers with NHERF2 cDNA in which its binding domains had been deleted. When only CFTR is expressed on the apical membrane, deletion of any of the NHERF2 binding domains inhibited both PKA-dependent apical CFTR expression and its activation, while when NHE3 was co-expressed with CFTR PDZ2 deletion was without effect on CFTR sorting and activity. This suggests that when the PDZ2 domain is "sequestered" by interacting with NHE3 it can no longer participate in CFTR functional expression.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cytoskeletal Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Colforsin/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Mutation , Nephrons/cytology , Nephrons/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/physiology , Transfection , Xenopus laevis
3.
Pflugers Arch ; 449(1): 66-75, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15235914

ABSTRACT

Nucleotide binding to purinergic P2Y receptors contributes to the regulation of a variety of physiological functions in renal epithelial cells. Here, we investigate the regulatory mechanism of the P2Y1 receptor agonist 2-methylthioadenosine diphosphate (2-MeSADP) on Cl- transport in A6 cells, a commonly used model of the distal section of the Xenopus laevis nephron. Protein and mRNA expression analysis together with functional measurements demonstrated the basolateral location of the Xenopus P2Y1 receptor. 2-MeSADP increased intracellular [Ca2+] and cAMP and Cl- efflux, responses that were all inhibited by the specific P2Y1 receptor antagonist MRS 2179. Cl- efflux was also inhibited by the cystic fibrosis transmembrane conductance regulator (CFTR) blocker glibenclamide. Inhibition of either protein kinase A (PKA) or the binding between A-kinase-anchoring proteins (AKAPs) and the regulatory PKA RII subunit blocked the 2-MeSADP-induced activation of CFTR, suggesting that PKA mediates P2Y1 receptor regulation of CFTR through one or more AKAPs. Further, the truncation of the PDZ1 domain of the scaffolding protein Na+/H+ exchanger regulatory factor-2 (NHERF-2) inhibited 2-MeSADP-dependent stimulation of Cl- efflux, suggesting the involvement of this scaffolding protein. Activation or inhibition of PKC had no effect per se on basal Cl- efflux but potentiated or reduced the 2-MeSADP-dependent stimulation of Cl- efflux, respectively. These data suggest that the X laevis P2Y1 receptor in A6 cells can increase both cAMP/PKA and Ca2+/PKC intracellular levels and that the PKC pathway is involved in CFTR activation via potentiation of the PKA pathway.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Phosphoproteins/metabolism , Receptors, Purinergic P2/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/metabolism , Animals , Calcium/metabolism , Cell Line , Chlorides/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/cytology , Glyburide/metabolism , Indomethacin/metabolism , Isoquinolines/metabolism , Kidney Tubules/cytology , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y1 , Sodium-Hydrogen Exchangers , Sulfonamides/metabolism , Thionucleotides/metabolism , Xenopus laevis
4.
Mol Reprod Dev ; 63(2): 210-22, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12203831

ABSTRACT

Bovine cumulus-oocyte complexes (COCs) and mural granulosa cells express the mRNA coding for the micro-opioid receptor. The addition of beta-endorphin (beta-end) to oocytes cultured in hormonally-supplemented in vitro maturation (IVM) medium had no effect on the rates of oocytes reaching the metaphase II (MII) stage, but significantly decreased the maturation rate (P < 0.05) and arrested oocytes at metaphase I (MI) after culture in hormone-free medium (P < 0.001). Naloxone (Nx) reverted this inhibitory effect of beta-end. Moreover, Nx "per se" showed a dose-dependent dual effect. When added at high concentration (10 x (-3) M), it significantly reduced the rate of oocytes in MII (P < 0.001), thus increasing the rate of oocytes arrested in MI. However, Nx added at low concentration (10 x (-8) M) significantly increased oocyte maturation (P < 0.001). High concentration of Nx induced an increase in both intracellular calcium concentration ([Ca(2+)](i)) and in the activity of the mitogen-activated protein kinase (MAPK) also called extracellular-regulated kinase (ERK) in cumulus cells of bovine COCs. Blocking the rise in [Ca(2+)](i) with the calcium chelator acetoxymethylester-derived form of bis (o-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM) reversed the Nx-dependent inhibition of meiotic maturation observed at high Nx concentrations. Whereas blocking ERK with the MAPK/ERK kinase (MEK) inhibitor, PD98059, had no effect on this process. Therefore, we concluded that the mocro-opioid receptor, by inducing [Ca(2+)](i) increase, participates in the cumulus-oocyte coupled signaling associated with oocyte maturation.


Subject(s)
Calcium/metabolism , Cell Differentiation/drug effects , Egtazic Acid/analogs & derivatives , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oocytes/drug effects , beta-Endorphin/pharmacology , Animals , Cattle , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Granulosa Cells/drug effects , In Vitro Techniques , Meiosis/drug effects , Mitogen-Activated Protein Kinases/drug effects , Oocytes/enzymology , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid, mu/genetics
5.
J Membr Biol ; 188(3): 249-59, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12181615

ABSTRACT

As potential autocrine or paracrine factors, extracellular nucleotides are known to be important regulators of renal ion transporters by activating cell surface receptors and intracellular signaling pathways. We investigated the influence of extracellular adenine nucleotides on Na+/H+ exchanger isoform 3 (NHE3) activity in A6-NHE3 cells. This is a polarized cell line obtained by stable transfection of A6 cells with the cDNA encoding the rat isoform of NHE3, which is expressed on the apical membrane. Basolateral addition of the P2Y(1) agonist, 2-MeSADP, induced an inhibition of NHE3 activity, which was prevented by preincubation with selective P2Y(1) antagonists, MRS 2179 (N6-methyl-2'-deoxyadenosine-3',5'-bisphosphate) and MRS 2286 (2-[2-(2-chloro-6-methylamino-purin-9-yl)-ethyl]-propane-1,3-bisoxy(diammoniumphosphate)). NHE3 activity was also significantly inhibited by ATP and ATP-gamma-S but not by UTP. 2-MeSADP induced a P2Y(1) antagonist-sensitive increase in both [Ca2+]i and cAMP production. Pre-incubation with a PKC inhibitor, Calphostin C, or the calcium chelator BAPTA-AM, had no effect on the 2-MeSADP-dependent inhibition of NHE3 activity, whereas this inhibition was reversed by either incubation with the PKA inhibitor H89 or by mutation of two PKA target serines (S552 and S605) on NHE3. Pre-incubation of the A6-NHE3 cells with the synthetic peptide, Ht31, which prevents the binding between AKAPs and the regulatory PKA subunits RII, also prevented the 2-MeSADP-induced inhibition of NHE3. We conclude that only the cAMP/PKA pathway is involved in the inhibition of NHE3 activity.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/metabolism , Calcium/metabolism , Cyclic AMP/biosynthesis , Epithelial Cells/physiology , Sodium-Hydrogen Exchangers/physiology , Thionucleotides/metabolism , Adenosine/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/physiology , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Hydrogen-Ion Concentration , Rats/genetics , Signal Transduction , Sodium-Hydrogen Exchangers/drug effects , Thionucleotides/pharmacology , Transfection , Xenopus laevis/genetics , Xenopus laevis/metabolism
6.
Exp Nephrol ; 9(5): 341-8, 2001.
Article in English | MEDLINE | ID: mdl-11549852

ABSTRACT

The opioid receptor antagonist, naloxone, has been shown to have beneficial effects in the kidney and to be implicated in renal salt and water balance. In the present study the signal transduction pathways utilized by naloxone were studied in an epithelial cell line model of the cortical collecting duct, A6 cells. We found that naloxone has a dual effect depending on the concentration used: at a low concentration (10(-6) M) it antagonized the beta-endorphin-dependent increase in cytoplasmic calcium [Ca(2+)](i), while at higher concentrations (>10(-5) M) it increased [Ca(2+)](i) and intracellular inositol phosphate levels. While naloxone-induced increases in [Ca(2+)](i) occurred in the absence of external calcium, it was significantly stimulated by increasing the external calcium concentration, suggesting that naloxone increases [Ca(2+)](i) via both calcium release and calcium influx. In polarized A6 cell monolayers naloxone inhibited the activity of the Na(+)/H(+) exchanger (NHE) only when added to the basolateral cell surface. This inhibition of the NHE was prevented by pretreatment of the cells with either the intracellular calcium chelator, BAPTA or with the protein kinase C inhibitor, calphostin C. These findings demonstrate that naloxone induces a rapid increase in intracellular calcium which inhibits the NHE via the calcium-dependent protein kinase C regulatory pathway.


Subject(s)
Calcium Signaling/drug effects , Egtazic Acid/analogs & derivatives , Naloxone/pharmacology , Protein Kinase C/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Calcium/metabolism , Calcium Signaling/physiology , Cell Line , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Naphthalenes/pharmacology , Narcotic Antagonists/pharmacology , Protein Kinase C/antagonists & inhibitors , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Spectrometry, Fluorescence , Sulfonamides/pharmacology , beta-Endorphin/metabolism
7.
J Membr Biol ; 178(2): 103-13, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11083899

ABSTRACT

We have previously demonstrated that in A(6) renal epithelial cells, a commonly used model of the mammalian distal section of the nephron, adenosine A(1) and A(2A) receptor activation modulates sodium and chloride transport and intracellular pH (Casavola et al., 1997). Here we show that apical addition of the A(3) receptor-selective agonist, 2-chloro-N(6)-(3-iodobenzyl)-adenosine-5'-methyluronamide (Cl-IB-MECA) stimulated a chloride secretion that was mediated by calcium- and cAMP-regulated channels. Moreover, in single cell measurements using the fluorescent dye Fura 2-AM, Cl-IB-MECA caused an increase in Ca(2+) influx. The agonist-induced rise in [Ca(2+)](i) was significantly inhibited by the selective adenosine A(3) receptor antagonists, 2,3-diethyl-4, 5-dipropyl-6-phenylpyridine-3-thiocarboxylate-5-carboxylate (MRS 1523) and 3-ethyl 5-benzyl 2-methyl-6-phenyl-4-phenylethynyl-1, 4-(+/-)-dihydropyridine-3,5-dicarboxylate (MRS 1191) but not by antagonists of either A(1) or A(2) receptors supporting the hypothesis that Cl-IB-MECA increases [Ca(2+)](i) by interacting exclusively with A(3) receptors. Cl-IB-MECA-elicited Ca(2+) entry was not significantly inhibited by pertussis toxin pretreatment while being stimulated by cholera toxin preincubation or by raising cellular cAMP levels with forskolin or rolipram. Preincubation with the protein kinase A inhibitor, H89, blunted the Cl-IB-MECA-elicited [Ca(2+)](i) response. Moreover, Cl-IB-MECA elicited an increase in cAMP production that was inhibited only by an A(3) receptor antagonist. Altogether, these data suggest that in A(6) cells a G(s)/protein kinase A pathway is involved in the A(3) receptor-dependent increase in calcium entry.


Subject(s)
Adenosine/analogs & derivatives , Calcium/metabolism , Chlorides/metabolism , Epithelial Cells/physiology , Kidney/physiology , Receptors, Purinergic P1/physiology , Adenosine/pharmacology , Animals , Calcium Signaling , Cell Line , Colforsin/pharmacology , Cyclic AMP/biosynthesis , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dihydropyridines/pharmacology , Electric Conductivity , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Kidney/drug effects , Kidney/metabolism , Phosphodiesterase Inhibitors/pharmacology , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Pyridines/pharmacology , Receptor, Adenosine A3 , Rolipram/pharmacology , Signal Transduction
8.
FASEB J ; 14(14): 2185-97, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053239

ABSTRACT

In this study we investigate the mechanism of intracellular pH change and its role in malignant transformation using the E7 oncogene of human papillomavirus type 16 (HPV16). Infecting NIH3T3 cells with recombinant retroviruses expressing the HPV16 E7 or a transformation deficient mutant we show that alkalinization is transformation specific. In NIH3T3 cells in which transformation can be turned on and followed by induction of the HPV16 E7 oncogene expression, we demonstrate that cytoplasmic alkalinization is an early event and was driven by stimulation of Na+/H+ exchanger activity via an increase in the affinity of the intracellular NHE-1 proton regulatory site. Annulment of the E7-induced cytoplasmic alkalinization by specific inhibition of the NHE-1, acidification of culture medium, or clamping the pHi to nontransformed levels prevented the development of later transformed phenotypes such as increased growth rate, serum-independent growth, anchorage-independent growth, and glycolytic metabolism. These findings were verified in human keratinocytes (HPKIA), the natural host of HPV. Results from both NIH3T3 and HPKIA cells show that alkalinization acts on pathways that are independent of the E2F-mediated transcriptional activation of cell cycle regulator genes. Moreover, we show that the transformation-dependent increase in proliferation is independent of the concomitant stimulation of glycolysis. Finally, treatment of nude mice with the specific inhibitor of NHE-1, DMA, delayed the development of HPV16-keratinocyte tumors. Our data confirm that activation of the NHE-1 and resulting cellular alkalinization is a key mechanism in oncogenic transformation and is necessary for the development and maintenance of the transformed phenotype.


Subject(s)
Amiloride/analogs & derivatives , Cell Transformation, Neoplastic , Sodium-Hydrogen Exchangers/physiology , 3T3 Cells , Amiloride/pharmacology , Animals , Binding Sites , Binding, Competitive , Cell Division/drug effects , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral/genetics , Culture Media, Serum-Free/pharmacology , Cyclin E/drug effects , Cyclin E/metabolism , Glycolysis , Humans , Hydrogen-Ion Concentration , Keratinocytes/cytology , Keratinocytes/virology , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/physiology , Papillomavirus E7 Proteins , Phenotype , S Phase , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Transplantation, Heterologous , Xenograft Model Antitumor Assays
9.
Pflugers Arch ; 440(6): 871-80, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11041553

ABSTRACT

HGT-1 is a human cell line sharing a number of physiological features with gastric parietal cells. HGT-1 cell monolayers were able to secrete H+ when stimulated with histamine (calculated external pH variation, deltapH(e) 0.46+/-0.05) as assessed using the impermeant, pH-sensitive fluorescence dye 8-hydroxypyrene-1,3,6-trisulphonic acid, trisodium salt (HPTS). Treatment with 100 microM omeprazole inhibited the histamine-induced apical acidification by about 60%. Intracellular pH (pH(i)) measurements using the fluorescent pH-sensitive dye 2',7'-bis-carboxyethyl-5(6)-carboxyfluorescein (BCECF) demonstrated the expression of a functional, omeprazole-sensitive H+/K+-pump. A monoclonal antibody directed against the alpha subunit of the H+/K+-ATPase immunoprecipitated a 95-kDa protein from HGT-1 cells and human stomach which corresponds to the expected molecular size of the native protein. HGT-1 cells were also positive for the anion exchanger AE2 that is expressed in gastric parietal cells. In addition, we identified a histamine- and pH(i)-sensitive Na+/H+ exchanger in HGT-1 cells, which might correspond to the functional expression of the NHE4 isoform that has been detected in gastric epithelial cells as well as in primary cultured parietal cells. HGT-1 cells therefore display the principal features of parietal cells and might represent an interesting cell culture model for studying the regulatory mechanisms involved in acid secretion.


Subject(s)
Gastric Acid/metabolism , Parietal Cells, Gastric/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Amiloride/pharmacology , Antibodies, Monoclonal , Cell Line , Coloring Agents , Enzyme Inhibitors/pharmacology , Fluoresceins , Fluorescent Antibody Technique , H(+)-K(+)-Exchanging ATPase/immunology , H(+)-K(+)-Exchanging ATPase/metabolism , Histamine/pharmacology , Humans , Hydrogen-Ion Concentration , Immunosorbent Techniques , Kinetics , Models, Biological , Omeprazole/pharmacology , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/ultrastructure , Proton Pump Inhibitors , Sodium/pharmacology , Tight Junctions/ultrastructure
10.
J Biol Chem ; 275(8): 5361-9, 2000 Feb 25.
Article in English | MEDLINE | ID: mdl-10681510

ABSTRACT

Whereas the tumor acidic extracellular pH plays a crucial role in the invasive process, the mechanism(s) behind this acidification, especially in low nutrient conditions, are unclear. The regulation of the Na(+)/H(+) exchanger (NHE) and invasion by serum deprivation were studied in a series of breast epithelial cell lines representing progression from non-tumor to highly metastatic cells. Whereas serum deprivation reduced lactate production in all three cells lines, it inhibited NHE activity in the non-tumor cells and stimulated it in the tumor cells with a larger stimulation in the metastatic cells. The stimulation of NHE in the tumor cell lines was the result of an increased affinity of the internal H(+) regulatory site of the NHE without changes in sodium kinetics or expression. Serum deprivation conferred increased cell motility and invasive ability that were abrogated by specific inhibition of the NHE. Inhibition of phosphoinositide 3-kinase by overexpression of a dominant-negative mutant or wortmannin incubation inhibited NHE activity and invasion in serum replete conditions while potentiating the serum deprivation-dependent activation of the NHE and invasion. These results indicate that the up-regulation of the NHE by a phosphoinositide 3-kinase-dependent mechanism plays an essential role in increased tumor cell invasion induced by serum deprivation.


Subject(s)
Blood , Breast Neoplasms/metabolism , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/physiology , Sodium-Hydrogen Exchangers/metabolism , Adenosine Triphosphate/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Ammonium Chloride/pharmacology , Androstadienes/pharmacology , Blotting, Western , Cell Line , Cell Movement , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Kinetics , Lactic Acid/biosynthesis , Phosphoinositide-3 Kinase Inhibitors , Sodium-Hydrogen Exchangers/genetics , Time Factors , Transfection , Tumor Cells, Cultured , Up-Regulation , Wortmannin
11.
Int J Oncol ; 16(1): 155-60, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10601561

ABSTRACT

Transforming growth factor-beta (TGF-beta) exerts an inhibitory effect on epithelial cell proliferation while insulin-like growth factor-1 (IGF-1) is a positive regulator of proliferation and together they may participate in driving neoplastic progression. The regulation of TGF-beta1 and IGF-1 gene expression was analyzed in an in vitro model of an estrogen receptor positive (ER+), non-metastatic (MCF-7) and an (ER-), metastatic (MDA-MB-435) breast cancer cell line, respectively. Our results indicate a loss of the regulation of TGF-beta1 and the gain of the expression and upregulation of IGF-1 pathways during malignant progression. These data demonstrate that two factors, convergent on cell growth, can have divergent roles in the regulation of the expression of TGF-beta1.


Subject(s)
Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor I/genetics , Neoplasm Metastasis , Transforming Growth Factor beta/genetics , Blood , Breast Neoplasms , Cell Division/physiology , Disease Progression , Estradiol/physiology , Feedback , Humans , Insulin-Like Growth Factor I/biosynthesis , Transforming Growth Factor beta/biosynthesis , Tumor Cells, Cultured
12.
Cell Prolif ; 32(2-3): 107-18, 1999.
Article in English | MEDLINE | ID: mdl-10535357

ABSTRACT

Although cytosolic expression of the protein pS2 (TFF1) is considered to be a marker of oestrogen receptor (OR) function, there exists some clinical data to suggest an inverse relationship of cytosolic pS2 to tumour proliferation. Although secreted from breast cancer cells, the relationship of pS2 secretion to tumour natural history has been little studied. The mechanisms and kinetics of pS2 release and its relation to tumour cell proliferation were studied in a human breast cancer cell line MCF-7 and verified in a preliminary clinical study. Stimulation by stripped serum or oestradiol resulted in parallel increases of proliferation and pS2 release in both time course and dose-response experiments. Direct pharmacological alterations of proliferation were followed by identical changes in pS2 release. The relationship between serum pS2 levels and tumour proliferative activity when analysed as a function of steroid status showed a slope of 0.56 in OR+ vs. 0.19 in OR- tumours. It is concluded that pS2 release from breast cancer cells is associated with their proliferation and measurement of serum pS2 levels might be a good predictor of tumour proliferative state and could permit noninvasive monitoring of this tumour parameter.


Subject(s)
Breast Neoplasms/pathology , Growth Substances/physiology , Proteins/physiology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/blood , Breast Neoplasms/surgery , Cell Division/drug effects , Culture Media, Serum-Free , Deoxyadenosines/pharmacology , Female , Growth Substances/blood , Growth Substances/genetics , Humans , Kinetics , Middle Aged , Proteins/analysis , Proteins/genetics , Regression Analysis , Trefoil Factor-1 , Tumor Cells, Cultured , Tumor Suppressor Proteins
13.
J Physiol ; 515 ( Pt 3): 829-42, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10066908

ABSTRACT

1. Adenosine influences the vectorial transport of Na+ and HCO3- across kidney epithelial cells. However, its action on effector proteins, such as the Na+-H+ exchanger NHE3, an epithelial brush border isoform of the Na+-H+ exchanger (NHE) gene family, is not yet defined. 2. The present study was conducted in Xenopus laevis distal nephron A6 epithelia which express both an apical adenosine receptor of the A1 type (coupled to protein kinase C (PKC)) and a basolateral receptor of the A2 type (coupled to protein kinase A (PKA)). The untransfected A6 cell line expresses a single NHE type (XNHE) which is restricted to the basolateral membrane and which is activated by PKA. 3. A6 cell lines were generated which express exogenous rat NHE3. Measurements of side-specific pHi recovery from acid loads in the presence of HOE694 (an inhibitor with differential potency towards individual NHE isoforms) detected an apical resistant Na+-H+ exchange only in transfected cell lines. The sensitivity of the basolateral NHE to HOE694 was unchanged, suggesting that exogenous NHE3 was restricted to the apical membrane. 4. Stimulation of the apical A1 receptor with N 6-cyclopentyladenosine (CPA) inhibited both apical NHE3 and basolateral XNHE. These effects were mimicked by the addition of the protein kinase C (PKC) activator phorbol 12-myristate 13-acetate (PMA) and partially prevented by the PKC inhibitor calphostin C which also blocked the effect of PMA. 5. Stimulation of the basolateral A2 receptor with CPA inhibited apical NHE3 and stimulated basolateral XNHE. These effects were mimicked by 8-bromo-cAMP and partially prevented by the PKA inhibitor H89 which entirely blocked the effect of 8-bromo-cAMP. 6. In conclusion, CPA inhibits rat NHE3 expressed apically in A6 epithelia via both the apical PKC-coupled A1 and the basolateral PKA-coupled A2 adenosine receptors.


Subject(s)
Adenosine/pharmacology , Epithelial Cells/physiology , Nephrons/physiology , Sodium-Hydrogen Exchangers/physiology , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Guanidines/pharmacology , Hydrogen-Ion Concentration , Protein Kinase C/metabolism , Quaternary Ammonium Compounds/pharmacology , Rats , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/physiology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics , Sulfones/pharmacology , Transfection , Xenopus laevis
14.
Kidney Int ; 53(5): 1269-77, 1998 May.
Article in English | MEDLINE | ID: mdl-9573542

ABSTRACT

The present study describes two Na+/H+ exchanger (NHE) isoforms in an immortalized rabbit renal cortical collecting tubule cell line (RC.SV3). Na+/H+ exchange activity was assayed using fluorescence measurements of intracellular pH (pHi) in monolayers mounted in a cuvette containing two fluid compartments, making it possible to independently measure Na+/H+ exchange activity on either the apical or basolateral surface. RC.SV3 monolayers express Na+/H+ exchange activities in both the apical and basolateral membrane domains. The two exchangers have half-saturation constants (Km) for external sodium and sensitivities to dimethylamiloride, to HOE-694 and to cimetidine and clonidine consistant with the NHE-1 isoform on the basolateral cell surface and the NHE-2 isoform on the apical surface. Protein kinase A inhibition of basolateral exchanger activity was significantly higher than that of the apical exchanger. Protein kinase C significantly stimulated both exchangers equally. RT-PCR analysis found RNA for only NHE-1 and NHE-2, and immunofluorescence with an antibody against NHE-1 demonstrated a basolateral location for this isoform. The results suggest that RC.SV3 cells have two Na+/H+ exchange activities separated spatially to the two cellular membranes, with the NHE-1 and the NHE-2 isoforms located on the basolateral and the apical membranes, respectively.


Subject(s)
Kidney Tubules, Collecting/metabolism , Sodium-Hydrogen Exchangers/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Base Sequence , Cell Line , Cell Membrane/metabolism , Cell Polarity , Cimetidine/pharmacology , Clonidine/pharmacology , DNA Primers/genetics , Guanidines/pharmacology , Hydrogen-Ion Concentration , Intracellular Fluid/metabolism , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Kinetics , Polymerase Chain Reaction , Protein Kinases/metabolism , Rabbits , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics , Sulfones/pharmacology
15.
FASEB J ; 11(10): 785-92, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9271363

ABSTRACT

Data concerning the hormone sensitivity of the release and role of the aspartyl protease cathepsin D in tumor proliferative and invasive processes have been contradictory. To clarify the mechanisms of its release and role we first studied the contribution of estradiol and stripped serum to the time course and kinetics of cathepsin D release, proliferation, and invasion in parallel in the MCF-7 in vitro breast cancer cell culture model. Both estradiol and stripped serum independently stimulated both proliferation and cathepsin D release. However, the dose-response of estradiol and stripped serum-dependent stimulated release were similar to those for invasion and differed from those for proliferation: cathepsin D release and invasion were first stimulated at a stripped serum concentration more than 10-fold lower than that which initiated proliferation and had half stimulation constants almost 10-fold lower than those for proliferation. These results demonstrate that cathepsin D release is not related in any direct way to proliferation. The effect of the reduction of cathepsin D activity or release on in vitro invasion was also measured: both the inhibition of secreted cathepsin D activity by a specific inhibitor, diazoacetyl-DL-Nle-OMe, and the reduction of cathepsin D release by antisense oligonucleotides against its translation start site reduced cellular in vitro invasion without affecting proliferation. Cathepsin D release and activity are concluded to be directly involved in the process of invasion.


Subject(s)
Breast Neoplasms/enzymology , Cathepsin D/physiology , Neoplasm Invasiveness/diagnosis , Blood , Breast Neoplasms/pathology , Cell Division , Culture Media , Estradiol/pharmacology , Female , Humans , Oligonucleotides, Antisense/pharmacology , Tumor Cells, Cultured
16.
Pflugers Arch ; 434(1): 123-31, 1997 May.
Article in English | MEDLINE | ID: mdl-9094265

ABSTRACT

NHE3 is most likely the isoform involved in renal reabsorption of HCO3- and Na+. The functional properties of the "cloned" NHE3 isoform, including its transport regulation by extra- and intracellular stimuli, have so far been studied using non-epithelial expression systems. In the present report we stably transfected NHE3 cDNA (rabbit isoform) into Madin-Darby canine kidney cells (MDCK) cells and compared the sensitivity to inhibitors and the regulation of the Na+/H+-exchanger by vasotocin in NHE3 transfectants to that of the intrinsic basolateral Na+/H+-exchanger in untransfected and control transfected MDCK cells. By Southern blot analysis we documented that the NHE3 transcript is expressed in NHE3 transfectants. Na+/H+-exchange activity, measured as sodium-dependent recovery of intracellular pH from an acid load using 2', 7'-bis(carboxymethyl)-5(6)-carboxy-fluorescein (BCECF), was equally present at the basolateral cell surface of all cell lines; however, NHE3 transfectants demonstrated transport activity in the apical membrane that was significantly higher than that in untransfected or control transfected MDCK cells. Studies with ethylisopropylamiloride (EIPA) have shown that there is a similar sensitivity to inhibitors of the apical and/or basolateral Na+/H+-exchanger in transfected and untransfected MDCK cell lines. In contrast, the apical Na+/H+-exchanger (as compared to the basolateral Na+/H+-exchanger) of NHE3 transfectants was found to be relatively insensitive to the inhibitor HOE 694. Vasotocin decreased the activity of the apical Na+/H+-exchanger in NHE3 transfectants and stimulated the activity of the basolateral Na+/H+-exchanger in transfected (with NHE3 or pMAMneo) and untransfected MDCK cells. Phorbol ester, as expected, increased the activity of the Na+/H+-exchanger in the basolateral membrane of all cell lines; also, it stimulated transport activity at the apical cell surface of NHE3 transfectants. No change of Na+/H+-exchange activities was seen in studies with 8-bromo-cAMP. The PKC inhibitor calphostin C completely suppressed regulation of the apical and/or basolateral Na+/H+-exchanger by vasotocin, it partially blocked activation of the apical Na+/H+-exchanger in NHE3 transfectants by phorbol 12-myristate 13-acetate (PMA), and completely blocked stimulation of basolateral Na+/H+-exchanger by PMA. Consistent with a V1 receptor action, the effects of vasotocin in NHE3 transfectants and in MDCK cells were blocked by the V1 receptor antagonist, d(CH2)5Tyr(Me)-AVP, but were not reproduced by the V2 receptor agonist desmopressin. It is concluded that NHE3 in the apical membrane of NHE3-transfected MDCK cells contributes to the differential regulation of the apical and basolateral Na+/H+-exchanger by vasotocin; NHE3 is inhibited and endogenous Na+/H+-exchange activity is stimulated by vasotocin via V1 receptor activation of the protein kinase C pathway.


Subject(s)
Kidney/drug effects , Sodium-Hydrogen Exchangers/drug effects , Vasotocin/pharmacology , Animals , Cells, Cultured , Dogs , Transfection
17.
Mol Pharmacol ; 51(3): 516-23, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9058608

ABSTRACT

The effect of adenosine on Na+/H+ exchange activity was examined in cultured A6 renal epithelial cells. Adenosine and its analogue N6-cyclopentyladenosine (CPA) had different effects on Na+/H+ exchange activity depending on the side of addition. Basolateral CPA induced a stimulation of Na+/H+ exchange activity that was completely prevented by preincubation with an A2A-selective antagonist, 8-(3-chlorostyryl)caffeine, whereas apical CPA induced a slight but significant inhibition of Na+/H+ exchange activity that was significantly reduced by the A1-receptor antagonist 1,3-dipropyl-8-cyclopentylxanthine. Protein kinase C activation may be involved in mediating the apical CPA inhibition of Na+/H+ exchange activity; this inhibition was prevented by the protein kinase C inhibitor calphostin C. Treatment with either forskolin or 8-bromo-cAMP significantly stimulated Na+/H+ exchange activity; only basolateral CPA addition induced an increase in cAMP level. These observations together with the finding that the CPA-dependent stimulation of exchange activity was prevented by the protein kinase A inhibitor H-89 support the hypothesis that basolateral CPA stimulates Na+/H+ exchange via adenylate cyclase/protein kinase A activation. Basolateral CPA also increased transepithelial Na+ transport, and this stimulation was prevented by the Na+/H+ exchange inhibitor HOE-694, suggesting that changes in pHi during hormone action can act as an intermediate in the second-messenger cascade.


Subject(s)
Adenosine/physiology , Hydrogen-Ion Concentration , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cell Line , Cyclic AMP/metabolism , Hydrogen/metabolism , Kidney , Purinergic P1 Receptor Antagonists , Sodium/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Xenopus laevis
18.
J Membr Biol ; 151(3): 237-45, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8661511

ABSTRACT

The effect of adenosine regulation on sodium and chloride transport was examined in cultured A6 renal epithelial cells. Adenosine and its analogue N6-cyclopentyladenosine (CPA) had different effects on short-circuit current (Isc) depending on the side of addition. Basolateral CPA addition induced an approximately threefold increase of the Isc that reached a maximum effect 20 min after addition and was completely inhibited by preincubation with either an A2 selective antagonist, CSC, or the sodium channel blocker, amiloride. Apical CPA addition induced a biphasic Isc response characterized by a rapid fourfold transient increase over its baseline followed by a decline and a plateau phase that were amiloride insensitive. The A1 adenosine antagonist, CPX, completely prevented this response. This Isc response to apical CPA was also strongly reduced in Cl--free media and was significantly inhibited either by basolateral bumetanide or apical DPC preincubation. Only basolateral CPA addition was able to induce an increase in cAMP level. CPA, added to cells in suspension, caused a rapid rise in [Ca2+]i that was antagonized by CPX, not affected by CSC and prevented by thapsigargin preincubation. These data suggest that basolateral CPA regulates active sodium transport via A2 adenosine receptors stimulating adenylate cyclase while apical CPA regulates Cl- secretion via A1 receptor-mediated changes in [Ca2+]i.


Subject(s)
Adenosine/physiology , Chlorides/metabolism , Receptors, Purinergic P1/analysis , Signal Transduction/physiology , Sodium/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Amiloride/pharmacology , Bumetanide/pharmacology , Calcium/analysis , Cell Line , Cell Membrane/metabolism , Chloride Channels/physiology , Cyclic AMP/metabolism , Diuretics/pharmacology , Dose-Response Relationship, Drug , In Vitro Techniques , Kidney/cytology , Receptors, Purinergic P1/drug effects , Signal Transduction/drug effects
19.
Am J Physiol ; 269(1 Pt 1): C226-33, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7543243

ABSTRACT

The selective A1-adenosine-receptor antagonist, 8-cyclopentyl-1,3-dipropylxanthine (CPX), has been reported to activate Cl- efflux from cystic fibrosis cells, such as pancreatic CFPAC-1 and lung IB3 cells bearing the cystic fibrosis transmembrane regulator(delta F508) mutation, but has little effect on the same process in cells repaired by transfection with wild-type cystic fibrosis transmembrane regulator (O. Eidelman, C. Guay-Broder, P. J. M. van Galen, K. A. Jacobson, C. Fox, R. J. Turner, Z. I. Cabantchik, and H. B. Pollard. Proc. Natl. Acad. Sci. USA 89: 5562-5566, 1992). We report here that CPX downregulates Na+/H+ exchange activity in CFPAC-1 cells but has a much smaller effect on cells repaired with the wild-type gene. CPX also mildly decreases resting intracellular pH. In CFPAC-1 cells, this downregulation is dependent on the presence of adenosine, since pretreatment of the cells with adenosine deaminase blocks the CPX effect. We also show that, by contrast, CPX action on these cells does not lead to alterations in intracellular free Ca2+ concentration. We conclude that CPX affects pH regulation in CFPAC-1 cells, probably by antagonizing the tonic action of endogenous adenosine.


Subject(s)
Cystic Fibrosis/metabolism , Hydrogen-Ion Concentration , Intracellular Membranes/metabolism , Purinergic P1 Receptor Antagonists , Xanthines/pharmacology , Calcium/metabolism , Cell Line , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator , Humans , Membrane Proteins/genetics , Osmolar Concentration , Pancreas/cytology , Pancreas/metabolism , Sodium-Hydrogen Exchangers/metabolism , Transfection
20.
J Membr Biol ; 135(3): 209-16, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8271260

ABSTRACT

We have used a well-differentiated A6-cell preparation (A6-C1) to study cellular location and vasopressin control of Na/H-exchange activity. After cell acidification, cell pHi (measured by BCECF-fluorescence) only recovered by the addition of Na medium to the basolateral cell surface; this pHi recovery was inhibited by dimethylamiloride (2 microM) consistent with basolateral location of Na/H-exchange activity. Addition of vasopressin produced stimulation of Na/H-exchange activity and increased the affinity of the exchanger for Na+. Stimulation of Na/H exchange was mimicked by pharmacological activation of protein kinase A (forskolin, 8-Br-cAMP) and not by pharmacological activation of protein kinase C (TPA). It is concluded that basolaterally located Na/H-exchange in A6-C1 cells is activated by vasopressin.


Subject(s)
Kidney Tubules/cytology , Sodium-Hydrogen Exchangers/physiology , Vasopressins/pharmacology , Animals , Cell Differentiation/physiology , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Epithelial Cells , Epithelium/chemistry , Epithelium/physiology , Fluorescence , Hydrogen-Ion Concentration , Kidney Tubules/chemistry , Kidney Tubules/physiology , Sodium-Hydrogen Exchangers/analysis , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...