Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters











Publication year range
1.
Am J Physiol Cell Physiol ; 313(1): C108-C117, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28515087

ABSTRACT

FXYD5 is a Na+-K+-ATPase regulator, expressed in a variety of normal epithelia. In parallel, it has been found to be associated with several types of cancer and effect lethal outcome by promoting metastasis. However, the molecular mechanism underlying FXYD5 mediated invasion has not yet been identified. In this study, using in vivo 4T1 murine breast cancer model, we found that FXYD5-specific shRNA significantly inhibited lung cancer metastasis, without having a substantial effect on primary tumor growth. Our study reveals that FXYD5 participates in multiple stages of metastatic development and exhibits more than one mode of E-cadherin regulation. We provide the first evidence that FXYD5-related morphological changes are mediated through its interaction with Na+-K+-ATPase. Experiments in cultured 4T1 cells have indicated that FXYD5 expression may downregulate the ß1 isoform of the pump. This behavior could have implications on both transcellular interactions and intracellular events. Further studies suggest that differential localization of the adaptor protein Annexin A2 in FXYD5-expressing cells may correlate with matrix metalloproteinase 9 secretion and adhesion changes in 4T1 wild-type cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Mammary Neoplasms, Experimental/genetics , Membrane Proteins/genetics , Neoplasms, Adipose Tissue/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Animals , Annexin A2/genetics , Annexin A2/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Female , Ion Channels , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Microfilament Proteins , Neoplasms, Adipose Tissue/metabolism , Neoplasms, Adipose Tissue/pathology , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sodium-Potassium-Exchanging ATPase/metabolism
2.
J Biol Chem ; 291(21): 11072-82, 2016 May 20.
Article in English | MEDLINE | ID: mdl-27006401

ABSTRACT

The FXYD proteins are a family of small membrane proteins that share an invariant four amino acid signature motif F-X-Y-D and act as tissue-specific regulatory subunits of the Na,K-ATPase. FXYD5 (also termed dysadherin or RIC) is a structurally and functionally unique member of the FXYD family. As other FXYD proteins, FXYD5 specifically interacts with the Na,K-ATPase and alters its kinetics by increasing Vmax However, unlike other family members FXYD5 appears to have additional functions, which cannot be readily explained by modulation of transport kinetics. Knockdown of FXYD5 in MDA-MB-231 breast cancer cells largely decreases expression and secretion of the chemokine CCL2 (MCP-1). A related effect has also been observed in renal cell carcinoma cells. The current study aims to further characterize the relationship between the expression of FXYD5 and CCL2 secretion. We demonstrate that transfection of M1 epithelial cell line with FXYD5 largely increases lipopolysaccharide (LPS) stimulated CCL2 mRNA and secretion of the translated protein. We have completed a detailed analysis of the molecular events leading to the above response. Our key findings indicate that FXYD5 generates a late response by increasing the surface expression of the TNFα receptor, without affecting its total protein level, or mRNA transcription. LPS administration to mice demonstrates induced secretion of CCL2 and TNFα in FXYD5-expressing lung peripheral tissue, which suggests a possible role for FXYD5 in normal epithelia during inflammation.


Subject(s)
Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Epithelial Cells/metabolism , Gene Expression/drug effects , Gene Knockdown Techniques , Humans , Inflammation Mediators/metabolism , Ion Channels , Kinetics , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Microfilament Proteins , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects
3.
Am J Physiol Cell Physiol ; 309(2): C126-35, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25994790

ABSTRACT

The Na+-K+-ATPase is specifically inhibited by cardiac glycosides, some of which may also function as endogenous mammalian hormones. Previous studies using Xenopus oocytes, yeast cells, or purified isoforms demonstrated that affinities of various cardiac glycosides for three isoforms of the Na+-K+-ATPase (α1-α3ß1) may differ, a finding with potential clinical implication. The present study investigates isoform selectivity and effects of cardiac glycosides on cultured mammalian cells under more physiological conditions. H1299 cells (non-small cell lung carcinoma) were engineered to express only one α-isoform (α1, α2, or α3) by combining stable transfection of isoforms and silencing endogenous α1. Cardiac glycoside binding was measured by displacement of bound 3H-ouabain. The experiments confirm moderate α1/α3:α2 selectivity of ouabain, moderate α2:α1 selectivity of digoxin, and enhanced α2:α1 selectivity of synthetic derivatives (Katz A, Tal DM, Heller D, Haviv H, Rabah B, Barkana Y, Marcovich AL, Karlish SJD. J Biol Chem 289: 21153-21162, 2014). Relative α2:α1 selectivity of digoxin vs. ouabain was also manifested by enhanced internalization of α2 in response to digoxin. Cellular proliferation assays of H1299 cells confirmed the patterns of α2:α1 selectivity for ouabain, digoxin, and a synthetic derivative and reveal a crucial role of surface pump density on sensitivity to cardiac glycosides. Because cardiac glycosides are being considered as drugs for treatment of cancer, effects of ouabain on proliferation of 12 cancer and noncancer cell lines, with variable plasma membrane expression of α1, have been tested. These demonstrated that sensitivity to ouabain indeed depends linearly on the plasma membrane surface density of Na+-K+-ATPase irrespective of status, malignant or nonmalignant.


Subject(s)
Antineoplastic Agents/pharmacology , Cardiac Glycosides/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/enzymology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Antineoplastic Agents/metabolism , Binding, Competitive , Cardiac Glycosides/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Digoxin/metabolism , Digoxin/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Humans , Isoenzymes , Neoplasms/genetics , Neoplasms/pathology , Ouabain/metabolism , Ouabain/pharmacology , Protein Binding , RNA Interference , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Transfection
4.
Am J Physiol Cell Physiol ; 306(11): C1080-8, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24717576

ABSTRACT

FXYD5 (dysadherin or also called a related to ion channel, RIC) is a transmembrane auxiliary subunit of the Na(+)-K(+)-ATPase shown to increase its maximal velocity (Vmax). FXYD5 has also been identified as a cancer-associated protein whose expression in tumor-derived cell lines impairs cytoskeletal organization and increases cell motility. Previously, we have demonstrated that the expression of FXYD5 in M1 cells derived from mouse kidney collecting duct impairs the formation of tight and adherence junctions. The current study aimed to further explore effects of FXYD5 at a single cell level. It was found that in M1, as well as three other cell lines, FXYD5 inhibits transformation of adhered single cells from the initial radial shape to a flattened, elongated shape in the first stage of monolayer formation. This is also correlated to less ordered actin cables and fewer focal points. Structure-function analysis has demonstrated that the transmembrane domain of FXYD5, and not its unique extracellular segment, mediates the inhibition of change in cell shape. This domain has been shown before to be involved in the association of FXYD5 with the Na(+)-K(+)-ATPase, which leads to the increase in Vmax. Furthermore, specific transmembrane point mutations in FXYD5 that either increase or decrease its effect on cell elongation had a corresponding effect on the coimmunoprecipitation of FXYD5 with α Na(+)-K(+)-ATPase. These findings lend support to the possibility that FXYD5 affects cell polarization through its transmembrane domain interaction with the Na(+)-K(+)-ATPase. Yet interaction of FXYD5 with other proteins cannot be excluded.


Subject(s)
Cell Polarity/physiology , Membrane Proteins/physiology , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Cell Line , Cell Line, Tumor , HEK293 Cells , Humans , Ion Channels , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Mice , Microfilament Proteins
5.
J Biol Chem ; 289(2): 1049-59, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24275648

ABSTRACT

Internalization of the Na(+)/K(+)-ATPase (the Na(+) pump) has been studied in the human lung carcinoma cell line H1299 that expresses YFP-tagged α1 from its normal genomic localization. Both real-time imaging and surface biotinylation have demonstrated internalization of α1 induced by ≥100 nm ouabain which occurs in a time scale of hours. Unlike previous studies in other systems, the ouabain-induced internalization was insensitive to Src or PI3K inhibitors. Accumulation of α1 in the cells could be augmented by inhibition of lysosomal degradation but not by proteosomal inhibitors. In agreement, the internalized α1 could be colocalized with the lysosomal marker LAMP1 but not with Golgi or nuclear markers. In principle, internalization could be triggered by a conformational change of the ouabain-bound Na(+)/K(+)-ATPase molecule or more generally by the disruption of cation homeostasis (Na(+), K(+), Ca(2+)) due to the partial inhibition of active Na(+) and K(+) transport. Overexpression of ouabain-insensitive rat α1 failed to inhibit internalization of human α1 expressed in the same cells. In addition, incubating cells in a K(+)-free medium did not induce internalization of the pump or affect the response to ouabain. Thus, internalization is not the result of changes in the cellular cation balance but is likely to be triggered by a conformational change of the protein itself. In physiological conditions, internalization may serve to eliminate pumps that have been blocked by endogenous ouabain or other cardiac glycosides. This mechanism may be required due to the very slow dissociation of the ouabain·Na(+)/K(+)-ATPase complex.


Subject(s)
Endocytosis/drug effects , Lysosomes/metabolism , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Biological Transport/drug effects , Blotting, Western , Cell Line, Tumor , Cycloheximide/pharmacology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lysosomal-Associated Membrane Protein 1/metabolism , Microscopy, Confocal , Potassium/pharmacology , Protein Synthesis Inhibitors/pharmacology , Proteolysis/drug effects , Pyrimidines/pharmacology , Rats , Sodium-Potassium-Exchanging ATPase/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
6.
J Steroid Biochem Mol Biol ; 139: 78-87, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24139875

ABSTRACT

Screening female rat distal colon preparations for aldosterone-induced genes identified the Hsp90-binding immunophilin FKBP51 as a major aldosterone-induced mRNA and protein. Limited induction of FKBP51 was observed also in other aldosterone-responsive tissues such as kidney medulla and heart. Ex vivo measurements in colonic tissue have characterized time course, dose response and receptor specificity of the induction of FKBP51. FKBP51 mRNA and protein were strongly up regulated by physiological concentrations of aldosterone in a late (greater than 2.5h) response to the hormone. Maximal increase in FKBP51 mRNA requires aldosterone concentrations that are higher than those needed to fully occupy the mineralocorticoid receptor (MR). Yet, the response is fully inhibited by the MR antagonist spironolactone and not inhibited and even stimulated by the glucocorticoid receptor (GR) antagonist RU486. These and related findings cannot be explained by a simple activation and dimerization of either MR or GR but are in agreement with response mediated by an MR-GR heterodimer. Overexpression or silencing FKBP51 in the kidney collecting duct cell line M1 had little or no effect on the aldosterone-induced increase in transepithelial Na(+) transport.


Subject(s)
Aldosterone/physiology , Intestinal Mucosa/metabolism , Tacrolimus Binding Proteins/genetics , Transcriptional Activation , Active Transport, Cell Nucleus , Aldosterone/pharmacology , Animals , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cell Membrane Permeability , Cells, Cultured , Colon/cytology , Colon/metabolism , Electric Impedance , Epithelial Cells/drug effects , Epithelial Cells/physiology , Female , Intestinal Mucosa/cytology , Intramolecular Transferases/genetics , Intramolecular Transferases/metabolism , Kidney Tubules, Collecting/cytology , Mice , Mifepristone/pharmacology , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoids/pharmacology , Mineralocorticoids/physiology , Protein Stability , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Tacrolimus Binding Proteins/metabolism , Tissue Culture Techniques
7.
J Biol Chem ; 287(25): 21130-41, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22535957

ABSTRACT

FXYD proteins are a group of short single-span transmembrane proteins that interact with the Na(+)/K(+) ATPase and modulate its kinetic properties. This study characterizes intracellular trafficking of two FXYD family members, FXYD1 (phospholemman (PLM)) and FXYD7. Surface expression of PLM in Xenopus oocytes requires coexpression with the Na(+)/K(+) ATPase. On the other hand, the Na(+)/Ca(2+) exchanger, another PLM-interacting protein could not drive it to the cell surface. The Na(+)/K(+) ATPase-dependent surface expression of PLM could be facilitated by either a phosphorylation-mimicking mutation at Thr-69 or a truncation of three terminal arginine residues. Unlike PLM, FXYD7 could translocate to the cell surface of Xenopus oocytes independently of the coexpression of α1ß1 Na(+)/K(+) ATPase. The Na(+)/K(+) ATPase-independent membrane translocation of FXYD7 requires O-glycosylation of at least two of three conserved threonines in its ectodomain. Subsequent experiments in mammalian cells confirmed the role of conserved extracellular threonine residues and demonstrated that FXYD7 protein, in which these have been mutated to alanine, is trapped in the endoplasmic reticulum and Golgi apparatus.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Animals , Cell Line, Tumor , Cell Membrane/genetics , Endoplasmic Reticulum/genetics , Gene Expression , Glycosylation , Golgi Apparatus/genetics , Humans , Membrane Proteins/genetics , Oocytes/cytology , Oocytes/metabolism , Phosphoproteins/genetics , Protein Structure, Tertiary , Protein Transport/physiology , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Xenopus laevis
8.
Am J Physiol Renal Physiol ; 301(6): F1270-80, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21900457

ABSTRACT

FXYD5 (dysadherin or RIC) is a member of the FXYD family of single-span transmembrane proteins associated with the Na(+)-K(+)-ATPase. Several studies have demonstrated enhanced expression of FXYD5 during metastasis and effects on cell adhesion and motility. The current study examines effects of FXYD5 on the paracellular permeability in the mouse kidney collecting duct cell line M1. Expressing FXYD5 in these cells leads to a large decrease in amiloride-insensitive transepithelial electrical resistance as well as increased permeability to 4-kDa dextran. Impairment of cell-cell contact was also demonstrated by staining cells for the tight and adherence junction markers zonula occludens-1 and ß-catenin, respectively. This is further supported by large expansions of the interstitial spaces, visualized in electron microscope images. Expressing FXYD5 in M1 cells resulted in a decrease in N-glycosylation of ß1 Na(+)-K(+)-ATPase, while silencing it in H1299 cells had an opposite effect. This may provide a mechanism for the above effects, since normal glycosylation of ß1 plays an important role in cell-cell contact formation (Vagin O, Tokhtaeva E, Sachs G. J Biol Chem 281: 39573-39587, 2006).


Subject(s)
Kidney Tubules, Collecting/physiology , Membrane Proteins/physiology , Amiloride/pharmacology , Animals , Cell Communication/drug effects , Cell Communication/physiology , Cell Line , Dextrans/chemistry , Electric Impedance , Gene Silencing , Glycosylation , Ion Channels , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/ultrastructure , Membrane Proteins/analysis , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Microfilament Proteins , Permeability , Phosphoproteins/analysis , Sodium Channel Blockers/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Zonula Occludens-1 Protein , beta Catenin/analysis
9.
Biochemistry ; 50(18): 3736-48, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21449573

ABSTRACT

The human α(1)/His(10)-ß(1) isoform of the Na,K-ATPase has been expressed in Pichia pastoris, solubilized in n-dodecyl-ß-maltoside, and purified by metal chelate chromatography. The α(1)ß(1) complex spontaneously associates in vitro with the detergent-solubilized purified human FXYD1 (phospholemman) expressed in Escherichia coli. It has been confirmed that FXYD1 spontaneously associates in vitro with the α(1)/His(10)-ß(1) complex and stabilizes it in an active mode. The functional properties of the α(1)/His(10)-ß(1) and α(1)/His(10)-ß(1)/FXYD1 complexes have been investigated by fluorescence methods. The electrochromic dye RH421 which monitors binding to and release of ions from the binding sites has been applied in equilibrium titration experiments to determine ion binding affinities and revealed that FXYD1 induces an ∼30% increase of the Na(+)-binding affinity in both the E(1) and P-E(2) conformations. By contrast, it does not affect the affinities for K(+) and Rb(+) ions. Phosphorylation induced partial reactions of the enzyme have been studied as backdoor phosphorylation by inorganic phosphate and in kinetic experiments with caged ATP in order to evaluate the ATP-binding affinity and the time constant of the conformational transition, Na(3)E(1)-P → P-E(2)Na(3). No significant differences with or without FXYD1 could be detected. Rate constants of the conformational transitions Rb(2)E(1) → E(2)(Rb(2)) and E(2)(Rb(2)) → Na(3)E(1), investigated with fluorescein-labeled Na,K-ATPase, showed only minor or no effects of FXYD1, respectively. The conclusion from all these experiments is that FXYD1 raises the binding affinity of α(1)ß(1) for Na ions, presumably at the third Na-selective binding site. In whole cell expression studies FXYD1 reduces the apparent affinity for Na ions. Possible reasons for the difference from this study using the purified recombinant Na,K-ATPase are discussed.


Subject(s)
Gene Expression Regulation, Enzymologic , Membrane Proteins/physiology , Phosphoproteins/physiology , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium/chemistry , Adenosine Triphosphate/chemistry , Animals , Binding Sites , Cattle , Dose-Response Relationship, Drug , Humans , Ions , Kinetics , Membrane Proteins/chemistry , Phosphoproteins/chemistry , Pichia/metabolism , Protein Isoforms , Recombinant Proteins/chemistry , Serum Albumin/chemistry , Spectrometry, Fluorescence/methods
SELECTION OF CITATIONS
SEARCH DETAIL