Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Cell Physiol Biochem ; 38(3): 883-92, 2016.
Article in English | MEDLINE | ID: mdl-26909737

ABSTRACT

BACKGROUND/AIMS: The potential role of the two-pore domain potassium channel KCNK5 (also known as TASK-2 and K(2P)5.1) in activated T cell physiology has only recently been described. So far KCNK5 has been described to be up-regulated in T cells in multiple sclerosis patients and to be implicated in the volume regulatory mechanism regulatory volume decrease (RVD) in T cells. METHODS: We investigated the time-dependent expression pattern of KCNK5 in CD3/CD28 activated human T cells using qPCR and Western blotting and its role in RVD using a Coulter Counter. RESULTS: KCNK5 is highly up-regulated in CD3/CD28 activated T cells both at mRNA (after 24 h) and protein level (72 and 144 h), but despite this up-regulation the RVD response is inhibited. Furthermore, the swelling-activated Cl- permeability in activated T cells is strongly decreased, and the RVD inhibition is predominantly due to the decreased Cl- permeability. CONCLUSION: The up-regulated KCNK5 in activated human T cells does not play a volume regulatory role, due to decreased Cl- permeability. We speculate that the KCNK5 up-regulation might play a role in hyperpolarization of the cell membrane leading to increased Ca2+ influx and proliferation of T cells.


Subject(s)
Lymphocyte Activation , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , T-Lymphocytes/metabolism , Up-Regulation , CD28 Antigens/metabolism , CD3 Complex/pharmacology , Calcium/metabolism , Cell Size/drug effects , Chlorine/metabolism , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics
2.
Channels (Austin) ; 9(6): 380-96, 2015.
Article in English | MEDLINE | ID: mdl-26569161

ABSTRACT

Volume-regulated channels for anions (VRAC) / organic osmolytes (VSOAC) play essential roles in cell volume regulation and other cellular functions, e.g. proliferation, cell migration and apoptosis. LRRC8A, which belongs to the leucine rich-repeat containing protein family, was recently shown to be an essential component of both VRAC and VSOAC. Reduced VRAC and VSOAC activities are seen in drug resistant cancer cells. ANO1 is a calcium-activated chloride channel expressed on the plasma membrane of e.g., secretory epithelia. ANO1 is amplified and highly expressed in a large number of carcinomas. The gene, encoding for ANO1, maps to a region on chromosome 11 (11q13) that is frequently amplified in cancer cells. Knockdown of ANO1 impairs cell proliferation and cell migration in several cancer cells. Below we summarize the basic biophysical properties of VRAC, VSOAC and ANO1 and their most important cellular functions as well as their role in cancer and drug resistance.


Subject(s)
Cell Size , Chloride Channels/metabolism , Drug Resistance, Neoplasm , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , Anoctamin-1 , Apoptosis , Chloride Channels/genetics , Homeostasis , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics
3.
Philos Trans R Soc Lond B Biol Sci ; 369(1638): 20130109, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24493757

ABSTRACT

Multi-drug resistance (MDR) to chemotherapy is the major challenge in the treatment of cancer. MDR can develop by numerous mechanisms including decreased drug uptake, increased drug efflux and the failure to undergo drug-induced apoptosis. Evasion of drug-induced apoptosis through modulation of ion transporters is the main focus of this paper and we demonstrate how pro-apoptotic ion channels are downregulated, while anti-apoptotic ion transporters are upregulated in MDR. We also discuss whether upregulation of ion transport proteins that are important for proliferation contribute to MDR. Finally, we discuss the possibility that the development of MDR involves sequential and localized upregulation of ion channels involved in proliferation and migration and a concomitant global and persistent downregulation of ion channels involved in apoptosis.


Subject(s)
Apoptosis/physiology , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/physiology , Ion Channels/metabolism , Models, Biological , Neoplasms/physiopathology , Humans , Ion Transport , Neoplasms/metabolism
5.
J Physiol ; 591(24): 6127, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24339147

Subject(s)
Apoptosis , Cell Size , Animals , Humans
6.
Front Physiol ; 4: 233, 2013.
Article in English | MEDLINE | ID: mdl-24009588

ABSTRACT

The physiological function of epithelia is transport of ions, nutrients, and fluid either in secretory or absorptive direction. All of these processes are closely related to cell volume changes, which are thus an integrated part of epithelial function. Transepithelial transport and cell volume regulation both rely on the spatially and temporally coordinated function of ion channels and transporters. In healthy epithelia, specific ion channels/transporters localize to the luminal and basolateral membranes, contributing to functional epithelial polarity. In pathophysiological processes such as cancer, transepithelial and cell volume regulatory ion transport are dys-regulated. Furthermore, epithelial architecture and coordinated ion transport function are lost, cell survival/death balance is altered, and new interactions with the stroma arise, all contributing to drug resistance. Since altered expression of ion transporters and channels is now recognized as one of the hallmarks of cancer, it is timely to consider this especially for epithelia. Epithelial cells are highly proliferative and epithelial cancers, carcinomas, account for about 90% of all cancers. In this review we will focus on ion transporters and channels with key physiological functions in epithelia and known roles in the development of cancer in these tissues. Their roles in cell survival, cell cycle progression, and development of drug resistance in epithelial cancers will be discussed.

7.
J Gen Physiol ; 141(5): 585-600, 2013 May.
Article in English | MEDLINE | ID: mdl-23630341

ABSTRACT

Members of the TMEM16 (Anoctamin) family of membrane proteins have been shown to be essential constituents of the Ca(2+)-activated Cl(-) channel (CaCC) in many cell types. In this study, we have investigated the electrophysiological properties of mouse TMEM16F. Heterologous expression of TMEM16F in HEK293 cells resulted in plasma membrane localization and an outwardly rectifying ICl,Ca that was activated with a delay of several minutes. Furthermore, a significant Na(+) current was activated, and the two permeabilities were correlated according to PNa = 0.3 PCl. The current showed an EC50 of 100 µM intracellular free Ca(2+) concentration and an Eisenman type 1 anion selectivity sequence of PSCN > PI > PBr > PCl > PAsp. The mTMEM16F-associated ICl,Ca was abolished in one mutant of the putative pore region (R592E) but retained in two other mutants (K616E and R636E). The mutant K616E had a lower relative permeability to iodide, and the mutant R636E had an altered anion selectivity sequence (PSCN = PI = PBr = PCl > PAsp). Our data provide evidence that TMEM16F constitutes a Ca(2+)-activated anion channel or a pore-forming subunit of an anion channel with properties distinct from TMEM16A.


Subject(s)
Calcium/metabolism , Chloride Channels/metabolism , Membrane Proteins/metabolism , Phospholipid Transfer Proteins/metabolism , Animals , Anions/metabolism , Anoctamins , Cell Membrane/metabolism , Cell Membrane/physiology , Cells, Cultured , HEK293 Cells , Humans , Ion Transport/physiology , Membrane Potentials/physiology , Mice , Permeability , Sodium/metabolism
8.
Cell Physiol Biochem ; 31(2-3): 366-78, 2013.
Article in English | MEDLINE | ID: mdl-23485744

ABSTRACT

BACKGROUND/AIMS: Signalling via CysLT1 is involved in activation of volume sensitive K(+) channels and homologous desensitization of the LTD4 receptor impairs regulatory volume decrease (RVD). The aim is to illustrate the effect of mutation of putative PKC consensus phosphorylation sites in the CysLT1R on desensitization and RVD. METHODS: mCysLT1 contains 4 putative PKC consensus phosphorylation sites, and four mutants were created: Thr151Gly, Thr323Gly, Thr151Gly plus Thr323Gly, and Thr236Gly plus Ser243Gly. Functional mCysLT1 receptor activity after injection of in vitro transcribed cRNA into Xenopus laevis oocytes was visualized as a LTD4-evoked, Ca(2+)-activated Cl(-) currents recorded by two-electrode voltage clamp. RESULTS: Repetitive LTD4 administration (100 nM) desensitized the LTD4-evoked currents in oocytes expressing wild type CysLT1. Single mutations as well as the double mutation Thr236Gly plus Ser243Gly had no or a slight effect on the LTD4 induced desensitization. However, double mutation Thr323Gly plus Thr151Gly prevented the desensitization. As a functional consequence we find that inhibition of PKC accelerates RVD and prevents the inhibitory effect of LTD4-pretreatment on RVD in Ehrlich ascites tumour cells. CONCLUSION: These data indicate that simultaneous PKC-mediated phosphorylation at the 2(nd) inner loop (Thr(151)) and at the C-terminal domain (Thr(323)) leads to mCysLT1 receptor desensitization and abrogates the RVD response following osmotic cell swelling.


Subject(s)
Protein Kinase C/metabolism , Receptors, Leukotriene/metabolism , Animals , Carbazoles/pharmacology , Cell Size , Chloride Channels/metabolism , Leukotriene D4/pharmacology , Mice , Mutagenesis, Site-Directed , Oocytes/metabolism , Patch-Clamp Techniques , Phosphorylation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Kinase C/antagonists & inhibitors , Receptors, Leukotriene/genetics , Signal Transduction/drug effects , Xenopus laevis/growth & development , Xenopus laevis/metabolism
9.
Compr Physiol ; 2(3): 2037-61, 2012 Jul.
Article in English | MEDLINE | ID: mdl-23723032

ABSTRACT

Cell shrinkage is a hallmark and contributes to signaling of apoptosis. Apoptotic cell shrinkage requires ion transport across the cell membrane involving K(+) channels, Cl(-) or anion channels, Na(+)/H(+) exchange, Na(+),K(+),Cl(-) cotransport, and Na(+)/K(+)ATPase. Activation of K(+) channels fosters K(+) exit with decrease of cytosolic K(+) concentration, activation of anion channels triggers exit of Cl(-), organic osmolytes, and HCO3(-). Cellular loss of K(+) and organic osmolytes as well as cytosolic acidification favor apoptosis. Ca(2+) entry through Ca(2+)-permeable cation channels may result in apoptosis by affecting mitochondrial integrity, stimulating proteinases, inducing cell shrinkage due to activation of Ca(2+)-sensitive K(+) channels, and triggering cell-membrane scrambling. Signaling involved in the modification of cell-volume regulatory ion transport during apoptosis include mitogen-activated kinases p38, JNK, ERK1/2, MEKK1, MKK4, the small G proteins Cdc42, and/or Rac and the transcription factor p53. Osmosensing involves integrin receptors, focal adhesion kinases, and tyrosine kinase receptors. Hyperosmotic shock leads to vesicular acidification followed by activation of acid sphingomyelinase, ceramide formation, release of reactive oxygen species, activation of the tyrosine kinase Yes with subsequent stimulation of CD95 trafficking to the cell membrane. Apoptosis is counteracted by mechanisms involved in regulatory volume increase (RVI), by organic osmolytes, by focal adhesion kinase, and by heat-shock proteins. Clearly, our knowledge on the interplay between cell-volume regulatory mechanisms and suicidal cell death is still far from complete and substantial additional experimental effort is needed to elucidate the role of cell-volume regulatory mechanisms in suicidal cell death.


Subject(s)
Apoptosis , Ion Transport , Animals , Humans , Ion Channels/metabolism , Ion Pumps/metabolism , MAP Kinase Signaling System , Potassium/metabolism
10.
Cell Physiol Biochem ; 28(6): 1231-46, 2011.
Article in English | MEDLINE | ID: mdl-22179011

ABSTRACT

Caveolae have been implicated in sensing of cell volume perturbations, yet evidence is still limited and findings contradictory. Here, we investigated the possible role of caveolae in cell volume regulation and volume sensitive signaling in an adipocyte system with high (3T3-L1 adipocytes); intermediate (3T3-L1 pre-adipocytes); and low (cholesterol-depleted 3T3-L1 pre-adipocytes) caveolae levels. Using large-angle light scattering, we show that compared to pre-adipocytes, differentiated adipocytes exhibit several-fold increased rates of volume restoration following osmotic cell swelling (RVD) and osmotic cell shrinkage (RVI), accompanied by increased swelling-activated taurine efflux. However, caveolin-1 distribution was not detectably altered after osmotic swelling or shrinkage, and caveolae integrity, as studied by cholesterol depletion or expression of dominant negative Cav-1, was not required for either RVD or RVI in pre-adipocytes. The insulin receptor (InsR) localizes to caveolae and its expression dramatically increases upon adipocyte differentiation. In pre-adipocytes, InsR and its effectors focal adhesion kinase (FAK) and extracellular signal regulated kinase (ERK1/2) localized to focal adhesions and were activated by a 5 min exposure to insulin (100 nM). Osmotic shrinkage transiently inhibited InsR Y(146)-phosphorylation, followed by an increase at t=15 min; a similar pattern was seen for ERK1/2 and FAK, in a manner unaffected by cholesterol depletion. In contrast, cell swelling had no detectable effect on InsR, yet increased ERK1/2 phosphorylation. In conclusion, differentiated 3T3-L1 adipocytes exhibit greatly accelerated RVD and RVI responses and increased swelling-activated taurine efflux compared to pre-adipocytes. Furthermore, in pre-adipocytes, Cav-1/caveolae integrity is not required for volume regulation. Given the relationship between hyperosmotic stress and insulin signaling, the finding that cell volume regulation is dramatically altered upon adipocyte differentiation may be relevant for the understanding of insulin resistance and metabolic syndrome.


Subject(s)
Adipocytes/physiology , Caveolae/metabolism , Focal Adhesion Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor, Insulin/metabolism , Signal Transduction , 3T3-L1 Cells , Adipocytes/enzymology , Adipocytes/metabolism , Animals , Cell Differentiation , Cell Size , Cholesterol/metabolism , Mice , Osmotic Pressure , Phosphorylation
11.
J Am Soc Nephrol ; 22(9): 1587-97, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21852585

ABSTRACT

Perturbations of cellular and systemic osmolarity severely challenge the function of all organisms and are consequently regulated very tightly. Here we outline current evidence on how cells sense volume perturbations, with particular focus on mechanisms relevant to the kidneys and to extracellular osmolarity and whole body volume homeostasis. There are a variety of molecular signals that respond to perturbations in cell volume and osmosensors or volume sensors responding to these signals. The early signals of volume perturbation include integrins, the cytoskeleton, receptor tyrosine kinases, and transient receptor potential channels. We also present current evidence on the localization and function of central and peripheral systemic osmosensors and conclude with a brief look at the still limited evidence on pathophysiological conditions associated with deranged sensing of cell volume.


Subject(s)
Cell Size , Mechanotransduction, Cellular , Homeostasis , Kidney/cytology , Kidney/physiology , Macrophages/physiology , Osmolar Concentration
12.
Cell Physiol Biochem ; 27(2): 109-20, 2011.
Article in English | MEDLINE | ID: mdl-21325828

ABSTRACT

Hsp70 has the ability to enhance the recovery of stressed cells by its ability to catalyze the reassembly of damaged proteins. Such a chaperoning function is essential for the Hsp70-mediated protection against anoxic stress that causes protein denaturation. We have studied induction of both transcription and translation of Hsp70 during recovery from chemical anoxia and the role of the extracellular signal regulated kinase ERK2 in this induction of Hsp70. 10 mM azide for 30 minutes (chemical anoxia) significantly inhibited the activity of ERK2 (measured as phospho-ERK) but the ERK-2 activity is rapidly increased in a MEK-independen manner, when azide is washed out of the cells. Chemical anoxia and overnight recovery induced Hsp70 expression (analyzed by Western blotting) and this was inhibited by actinomycin D as well as by cycloheximide showing that induction of both translation and transcription was involved. Inhibition of the MAP kinase p38, which was transiently activated during chemical anoxia, had no effect on the increase in Hsp70 expression whereas an inhibitor of reactive oxygen species and inhibition of the phosphatase PP1 and PP2a inhibited the increase in Hsp70 expression. Inhibition of ERK2 by the MEK inhibitor PD98059 resulted in strong inhibition of Hsp70 protein expression and simultaneous stimulation of hsp70 transcription.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Animals , Cell Hypoxia , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Flavonoids/pharmacology , HSP70 Heat-Shock Proteins/genetics , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , NIH 3T3 Cells , Protein Biosynthesis , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Protein Synthesis Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Sodium Azide/pharmacology , Transcriptional Activation , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Cell Physiol Biochem ; 25(2-3): 279-92, 2010.
Article in English | MEDLINE | ID: mdl-20110689

ABSTRACT

Cell motility and migration play pivotal roles in numerous physiological and pathophysiological processes including development and tissue repair. Cell migration is regulated through external stimuli such as platelet-derived growth factor-AA (PDGF-AA), a key regulator in directional cell migration during embryonic development and a chemoattractant during postnatal migratory responses including wound healing. We previously showed that PDGFRalpha signaling is coordinated by the primary cilium in quiescent cells. However, little is known about the function of the primary cilium in cell migration. Here we used micropipette analysis to show that a normal chemosensory response to PDGF-AA in fibroblasts requires the primary cilium. In vitro and in vivo wound healing assays revealed that in ORPK mouse (IFT88(Tg737Rpw)) fibroblasts, where ciliary assembly is defective, chemotaxis towards PDGF-AA is absent, leading to unregulated high speed and uncontrolled directional cell displacement during wound closure, with subsequent defects in wound healing. These data suggest that in coordination with cytoskeletal reorganization, the fibroblast primary cilium functions via ciliary PDGFRalpha signaling to monitor directional movement during wound healing.


Subject(s)
Cell Movement , Chemotaxis/physiology , Cilia/physiology , Platelet-Derived Growth Factor/metabolism , Wound Healing/physiology , Animals , Cells, Cultured , Fibroblasts/metabolism , Mice , NIH 3T3 Cells , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
Physiol Rev ; 89(1): 193-277, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19126758

ABSTRACT

The ability to control cell volume is pivotal for cell function. Cell volume perturbation elicits a wide array of signaling events, leading to protective (e.g., cytoskeletal rearrangement) and adaptive (e.g., altered expression of osmolyte transporters and heat shock proteins) measures and, in most cases, activation of volume regulatory osmolyte transport. After acute swelling, cell volume is regulated by the process of regulatory volume decrease (RVD), which involves the activation of KCl cotransport and of channels mediating K(+), Cl(-), and taurine efflux. Conversely, after acute shrinkage, cell volume is regulated by the process of regulatory volume increase (RVI), which is mediated primarily by Na(+)/H(+) exchange, Na(+)-K(+)-2Cl(-) cotransport, and Na(+) channels. Here, we review in detail the current knowledge regarding the molecular identity of these transport pathways and their regulation by, e.g., membrane deformation, ionic strength, Ca(2+), protein kinases and phosphatases, cytoskeletal elements, GTP binding proteins, lipid mediators, and reactive oxygen species, upon changes in cell volume. We also discuss the nature of the upstream elements in volume sensing in vertebrate organisms. Importantly, cell volume impacts on a wide array of physiological processes, including transepithelial transport; cell migration, proliferation, and death; and changes in cell volume function as specific signals regulating these processes. A discussion of this issue concludes the review.


Subject(s)
Cell Size , Osmosis/physiology , Signal Transduction/physiology , Vertebrates/physiology , Animals , Cell Death/physiology , Cell Movement/physiology , Cell Proliferation , Humans
15.
Am J Physiol Cell Physiol ; 295(6): C1528-34, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18842831

ABSTRACT

The transcription factor, tonicity-responsive enhancer binding protein (TonEBP), is involved in the adaptive response against hypertonicity. TonEBP regulates the expression of genes that catalyze the accumulation of osmolytes, and its transcriptional activity is increased by hypertonicity. The goal of the present investigation was to investigate whether cell shrinkage or high intracellular ionic concentration induced the activation of TonEBP. We designed a model system for isotonically shrinking cells over a prolonged period of time. Cells swelled in hypotonic medium and performed a regulatory volume decrease. Upon return to the original isotonic medium, cells shrank initially, followed by a regulatory volume increase. To maintain cell shrinkage, the RVI process was inhibited as follows: ethyl-isopropyl-amiloride inhibited the Na(+)/H(+) antiport, bumetanide inhibited the Na(+)-K(+)-2Cl(-) cotransporter, and gadolinium inhibited shrinkage-activated Na(+) channels. Cells remained shrunken for at least 4 h (isotonically shrunken cells). The activity of TonEBP was investigated with a Luciferase assay after isotonic shrinkage and after shrinkage in a high-NaCl hypertonic medium. We found that TonEBP was strongly activated after 4 and 16 h in cells in high-NaCl hypertonic medium, but not after 4 or 16 h in isotonically shrunken cells. Cells treated with high-NaCl hypertonic medium for 4 h had significantly higher intracellular concentrations of both K(+) and Na(+) than isotonically shrunken cells. This strongly suggested that an increase in intracellular ionic concentration and not cell shrinkage is involved in TonEBP activation.


Subject(s)
Cytoplasm/chemistry , Gene Expression Regulation/physiology , NFATC Transcription Factors/biosynthesis , Water-Electrolyte Balance/physiology , Animals , Cell Size , Hypotonic Solutions , Mice , NFATC Transcription Factors/genetics , NIH 3T3 Cells , Osmotic Pressure/physiology , Transcription, Genetic
16.
Proteomics ; 8(10): 2035-44, 2008 May.
Article in English | MEDLINE | ID: mdl-18491317

ABSTRACT

In the literature, a variety of ways have been used to obtain anoxia, and most often results are compared between studies without taking into consideration how anoxia has been obtained. Here, we provide a comprehensive study of two types of anoxia, using a proteomics approach to compare changes in protein expression. The two investigated situations were 30 min of chemical anoxia (10 mM NaN(3)) followed by reoxygenation overnight (CR) and 2 h of N(2)-induced anoxia (achieved by flushing with N(2)) followed by reoxygenation overnight (NR), after which samples were resolved by 2-DE. Forty-five protein spots changed their abundance in response to CR and 35 protein spots changed their abundance in response to NR, but only six proteins changed their abundance in response to both stimuli. By the means of MS/MS, 40 protein spots were identified including proteins involved in processes like cell protection and protein synthesis. It was also revealed that the level of a number of keratins was down-regulated. This study therefore provides a valuable comparison of two different anoxia models and shows that great care should be taken when comparing the effects of anoxia in studies that have used different types and durations of anoxia.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Fish Proteins/analysis , Proteome/analysis , Proteomics/methods , Tandem Mass Spectrometry/methods , Animals , Cell Hypoxia , Cell Line , Down-Regulation , Keratins/analysis , Keratins/metabolism , Oncorhynchus mykiss
17.
Cell Physiol Biochem ; 21(5-6): 373-84, 2008.
Article in English | MEDLINE | ID: mdl-18453745

ABSTRACT

Several types of K(+) channels have been identified in epithelial cells. Among them high conductance Ca(2+)-activated K(+) channels (BK channels) are of relevant importance for their involvement in regulatory volume decrease (RVD) response following hypotonic stress. The aim of the present work was to investigate the functional and molecular expression of BK in the eel intestine, which is a useful experimental model for cell volume regulation research. In the present paper using rat BK channel-specific primer, a RT-PCR signal of 696 pb cDNA was detected in eel intestine, whole nucleotide sequence showed high similarity (83%) to the alpha subunit of BK channel family. BK channel protein expression was verified by immunoblotting and confocal microscopy, while the functional role of BK channels in epithelial ion transport mechanisms and cell volume regulation was examined by electrophysiological and morphometric analysis on the intact tissue. BK(Ca) channels appeared to be localized along all the plasma membrane of the enterocytes; the apical part of the villi showed the most intense immunostaining. These channels were silent in basal condition, but were activated on both membranes (apical and basolateral) by increasing intracellular Ca(2+) concentration with the Ca(2+) ionophore ionomycin (1 microM). BK(Ca) channels were also activated on both membranes by hypotonic swelling of the epithelium and their inhibition by 100 nM iberiotoxin (specific BK(Ca) inhibitor) abolished the Regulatory Volume Decrease (RVD) of the intestinal cells after hypotonic swelling. In conclusion, our results demonstrated the molecular and functional expression of high conductance Ca(2+) -activated K(+) channels in eel intestine; the physiological role of these channels is mainly related to the RVD response of the epithelial cells following hypotonic swelling.


Subject(s)
Eels/metabolism , Intestinal Mucosa/metabolism , Potassium Channels, Calcium-Activated/metabolism , Amino Acid Sequence , Animals , Humans , Intestinal Mucosa/chemistry , Microscopy, Confocal , Molecular Sequence Data , Potassium Channels, Calcium-Activated/chemistry , Potassium Channels, Calcium-Activated/genetics , Sequence Alignment , Sequence Homology, Amino Acid
18.
Proteomics ; 8(5): 1009-18, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18240135

ABSTRACT

Twenty-four hours of N(2) induced anoxia induced global perturbations on protein expression in rainbow trout hypodermal fibroblasts cell line. Anoxia was obtained by depleting the medium of O(2) by flushing with N(2), and protein changes were studied by 2-DE coupled with MS providing quantitative measurements of a large number of proteins in one single study. The anoxic insult changed the level of 33 protein spots: 22 of these were up-regulated compared to the control situation and 11 were down-regulated. Using MS/MS sequencing 19 of the 33 protein spots that changed were identified, corresponding to a success rate of more than 50%. The identified proteins included two proteins involved in energy metabolism namely phosphoglycerate mutase and isocitrate dehydrogenase. In addition we observed the up-regulation of a cluster of proteins that contribute to cytoskeleton function. These are calpain, EB1, and Rho GDP dissociation inhibitor (GDI). The up-regulation of Rho GDI was shown to develop in a time dependent manner with no significant increase for up to 8 h of anoxia. In conclusion, this study provides a thorough investigation of the effect of anoxia in a cell line from rainbow trout.


Subject(s)
Fibroblasts/chemistry , Fish Proteins/analysis , Hypoxia , Oncorhynchus mykiss/physiology , Proteomics , Animals , Cell Line , Electrophoresis, Gel, Two-Dimensional , Tandem Mass Spectrometry
19.
Pflugers Arch ; 455(6): 1055-62, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17952454

ABSTRACT

The expression of the H-ras oncogene increases the migratory activity of many cell types and thereby contributes to the metastatic behavior of tumor cells. Other studies point to an involvement of volume-activated anion channels (VRAC) in (tumor) cell migration. In this paper, we tested whether VRACs are required for the stimulation of cell migration upon expression of the H-ras oncogene. We compared VRAC activation and migration of wild-type and H-ras-transformed NIH3T3 fibroblasts by means of patch-clamp techniques and time-lapse video microscopy. Both cell types achieve the same degree of VRAC activation upon maximal stimulation, induced by reducing extracellular osmolarity from 300 to 190 mOsm/l. However, upon physiologically relevant reductions in extracellular osmolarity (275 mOsm/l), the level of VRAC activation is almost three times higher in H-ras-transformed compared to wild-type fibroblasts. This increase in VRAC sensitivity is accompanied by increased migratory activity of H-ras fibroblasts. Moreover, the high-affinity VRAC blocker NS3728 inhibits migration of H-ras fibroblasts dose-dependently by up to about 60%, whereas migration of wild-type fibroblasts is reduced by only about 35%. Consistent with higher VRAC activity in H-ras than in wild-type fibroblasts, more VRAC blocker is needed to achieve a comparable degree of inhibition of migration. We suggest that H-ras modulates the volume set point of VRAC and thus facilitates transient changes of cell volume required for faster cell migration.


Subject(s)
Cell Movement/physiology , Genes, ras/physiology , Ion Channel Gating/physiology , Animals , Carbanilides , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/genetics , Cell Survival/physiology , Dose-Response Relationship, Drug , Electrophysiology , Fibroblasts/drug effects , Genes, ras/genetics , Ion Channel Gating/genetics , Mice , NIH 3T3 Cells , Osmolar Concentration , Patch-Clamp Techniques , Transformation, Genetic/genetics , Transformation, Genetic/physiology , Urea/analogs & derivatives , Urea/pharmacology
20.
Am J Physiol Cell Physiol ; 294(1): C197-212, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17977945

ABSTRACT

Hyperosmotic shrinkage induces multiple cellular responses, including activation of volume-regulatory ion transport, cytoskeletal reorganization, and cell death. Here we investigated the possible roles of ezrin/radixin/moesin (ERM) proteins in these events. Osmotic shrinkage of Ehrlich Lettre ascites cells elicited the formation of long microvillus-like protrusions, rapid translocation of endogenous ERM proteins and green fluorescent protein-tagged ezrin to the cortical region including these protrusions, and Thr(567/564/558) (ezrin/radixin/moesin) phosphorylation of cortical ERM proteins. Reduced cell volume appeared to be the critical parameter in hypertonicity-induced ERM protein activation, whereas alterations in extracellular ionic strength or intracellular pH were not involved. A shrinkage-induced increase in the level of membrane-associated phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)] appeared to play an important role in ERM protein activation, which was prevented after PtdIns(4,5)P(2) depletion by expression of the synaptojanin-2 phosphatase domain. While expression of constitutively active RhoA increased basal ERM phosphorylation, the Rho-Rho kinase pathway did not appear to be involved in shrinkage-induced ERM protein phosphorylation, which was also unaffected by the inhibition or absence of Na(+)/H(+) exchanger isoform (NHE1). Ezrin knockdown by small interfering RNA increased shrinkage-induced NHE1 activity, reduced basal and shrinkage-induced Rho activity, and attenuated the shrinkage-induced formation of microvillus-like protrusions. Hyperosmolarity-induced cell death was unaltered by ezrin knockdown or after phosphatidylinositol 3-kinase (PI3K) inhibition. In conclusion, ERM proteins are activated by osmotic shrinkage in a PtdIns(4,5)P(2)-dependent, NHE1-independent manner. This in turn mitigates the shrinkage-induced activation of NHE1, augments Rho activity, and may also contribute to F-actin rearrangement. In contrast, no evidence was found for the involvement of an NHE1-ezrin-PI3K-PKB pathway in counteracting shrinkage-induced cell death.


Subject(s)
Carcinoma, Ehrlich Tumor/metabolism , Cell Membrane/metabolism , Cell Size , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Actins/metabolism , Animals , Bicarbonates/metabolism , COS Cells , Carcinoma, Ehrlich Tumor/enzymology , Carcinoma, Ehrlich Tumor/pathology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Death , Cell Membrane/enzymology , Chlorocebus aethiops , Cytoskeletal Proteins/genetics , Cytoskeleton/enzymology , LLC-PK1 Cells , Mice , NIH 3T3 Cells , Osmotic Pressure , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation , Protein Transport , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Saline Solution, Hypertonic/metabolism , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Swine , Time Factors , Transfection , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...