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










Publication year range
1.
Int J Mol Sci ; 24(21)2023 Oct 28.
Article in English | MEDLINE | ID: mdl-37958687

ABSTRACT

Melanoma is a highly aggressive type of skin cancer produced through the malignant transformation of melanocytes, and it is usually associated with a poor prognosis. Clinically, melanoma has several stages associated with migration and invasion of the cells through the skin's layers, the rapid spreading of cells and the formation of tumors in multiple organs. The main problem is the emergence of resistance in melanoma to the applied methods of treatment; thus, it is of primary importance to find more crucial signaling pathways that control the progression of this type of cancer and could be targeted to prevent melanoma spreading. Here, we uncover novel aspects of the role of the mechanosensitive ion channel Piezo1 in melanoma tumor formation. Using a combinative approach, we showed the functional expression of mechanosensitive Piezo1 channels in the aggressive human melanoma SK-MEL-2 cell line. We found that chemical activation of Piezo1 by its agonist, Yoda1, prevents melanoma spheroid formation; thus, Piezo1 could be a potential target for selective modulation aimed at the prevention of melanoma development.


Subject(s)
Ion Channels , Melanoma , Humans , Ion Channels/genetics , Ion Channels/metabolism , Melanoma/genetics , Signal Transduction
2.
Biochem Biophys Res Commun ; 687: 149187, 2023 12 20.
Article in English | MEDLINE | ID: mdl-37944472

ABSTRACT

Sodium influx carried out by ion channels is one of the main regulators of water-salt and volume balance in cells of blood origin. Previously, we described amiloride-insensitive ENaC-like channels in human myeloid leukemia K562 cells; the intracellular regulatory mechanisms of the channels are associated with actin cytoskeleton dynamics. Recently, an extracellular mechanism of ENaC-like channels activation in K562 cells by the action of serine protease trypsin has been revealed. The other extracellular pathways that modulate ENaC (epithelial Na+ channel) activity and sodium permeability in transformed blood cells are not yet fully investigated. Here, we study the action of capsazepine (CPZ), as δ-ENaC activator, on single channel activity in K562 cells in whole-cell patch clamp experiments. Addition of CPZ (2 µM) to the extracellular solution caused an activation of sodium channels with typical features; unitary conductance was 15.1 ± 0.8 pS. Amiloride derivative benzamil (50 µM) did not inhibit their activity. Unitary currents and conductance of CPZ-activated channels were higher in Na+-containing extracellular solution than in Li+, that is one of the main fingerprints of δ-ENaC. The results of RT-PCR analysis and immunofluorescence staining also confirmed the expression of δ-hENaC (as well as α-, ß-, γ-ENaC) at the mRNA and protein level. These findings allow us to speculate that CPZ activates amiloride-insensitive ENaC-like channels that contain δ-ENaC in К562 cells. Our data reveal a novel extracellular mechanism for ENaC-like activation in human leukemia cells.


Subject(s)
Amiloride , Leukemia, Myeloid , Humans , Amiloride/pharmacology , Amiloride/metabolism , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Leukemia, Myeloid/metabolism , Sodium/metabolism , Oocytes/metabolism
3.
Membranes (Basel) ; 13(6)2023 Jun 04.
Article in English | MEDLINE | ID: mdl-37367787

ABSTRACT

Calcium-activated potassium channels (KCa) are important participants in calcium signaling pathways due to their ability to be activated by an increase in intracellular free calcium concentration. KCa channels are involved in the regulation of cellular processes in both normal and pathophysiological conditions, including oncotransformation. Previously, using patch-clamp, we registered the KCa currents in the plasma membrane of human chronic myeloid leukemia K562 cells, whose activity was controlled by local Ca2+ entry via mechanosensitive calcium-permeable channels. Here, we performed the molecular and functional identification of KCa channels and have uncovered their role in the proliferation, migration and invasion of K562 cells. Using a combined approach, we identified the functional activity of SK2, SK3 and IK channels in the plasma membrane of the cells. Selective SK and IK channel inhibitors, apamin and TRAM-34, respectively, reduced the proliferative, migratory and invasive capabilities of human myeloid leukemia cells. At the same time, the viability of K562 cells was not affected by KCa channel inhibitors. Ca2+ imaging showed that both SK and IK channel inhibitors affect Ca2+ entry and this could underlie the observed suppression of pathophysiological reactions of K562 cells. Our data imply that SK/IK channel inhibitors could be used to slow down the proliferation and spreading of chronic myeloid leukemia K562 cells that express functionally active KCa channels in the plasma membrane.

4.
J Cell Physiol ; 238(5): 918-930, 2023 05.
Article in English | MEDLINE | ID: mdl-36947588

ABSTRACT

PIEZO1 is a mechanosensitive channel widely presented in eukaryotic organisms. Although the PIEZO family was discovered in 2010, main questions related to the molecular structure as well as to specific activation mechanisms and regulating pathways remain open. Two hypotheses of PIEZO1 gating were formulated: the first, as a dominant hypothesis, through the plasma membrane (force-from-lipids) and the second, via the participation of the cytoskeleton and the components of the extracellular matrix (ECM) (force-from-filaments). Many researchers provide convincing evidence for both hypotheses. It was demonstrated that PIEZO1 has a propeller-like shape forming a membrane curvature within the lipid bilayer. That suggests the participation of lipids in channel modulation, and many studies demonstrate the critical role of lipids and compounds that modify the lipid bilayer in the regulation of PIEZO1 properties. At the same time, the components of ECM and cortical cytoskeleton can be affected by the membrane curvature and thus have an impact on PIEZO1 properties. In living cells, PIEZO1 properties are reported to be critically dependent on channel microenvironment that is on combinatorial influence of plasma membrane, cytoskeleton and ECM. Thus, it is necessary to understand which factors can affect PIEZO1 and consider them when interpreting the role of PIEZO1 in various physiological processes. This review summarizes the current knowledge about regulation of Piezo1 by lipids and the components of ECM and cytoskeleton.


Subject(s)
Ion Channel Gating , Ion Channels , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Lipid Bilayers/metabolism , Mechanotransduction, Cellular , Humans
5.
Front Oncol ; 12: 904742, 2022.
Article in English | MEDLINE | ID: mdl-35837090

ABSTRACT

Lung cancer is one of the most common cancer types in the world. Despite existing treatment strategies, overall patient survival remains low and new targeted therapies are required. Acidification of the tumor microenvironment drives the growth and metastasis of many cancers. Acid sensors such as acid-sensing ion channels (ASICs) may become promising targets for lung cancer therapy. Previously, we showed that inhibition of the ASIC1 channels by a recombinant analogue of mambalgin-2 from Dendroaspis polylepis controls oncogenic processes in leukemia, glioma, and melanoma cells. Here, we studied the effects and molecular targets of mambalgin-2 in lung adenocarcinoma A549 and Lewis cells, lung transformed WI-38 fibroblasts, and lung normal HLF fibroblasts. We found that mambalgin-2 inhibits the growth and migration of A549, metastatic Lewis P29 cells, and WI-38 cells, but not of normal fibroblasts. A549, Lewis, and WI-38 cells expressed different ASIC and ENaC subunits, while normal fibroblasts did not at all. Mambalgin-2 induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma cells. In line, acidification-evoked inward currents were observed only in A549 and WI-38 cells. Gene knockdown showed that the anti-proliferative and anti-migratory activity of mambalgin-2 is dependent on the expression of ASIC1a, α-ENaC, and γ-ENaC. Using affinity extraction and immunoprecipitation, mambalgin-2 targeting of ASIC1a/α-ENaC/γ-ENaC heteromeric channels in A549 cells was shown. Electrophysiology studies in Xenopus oocytes revealed that mambalgin-2 inhibits the ASIC1a/α-ENaC/γ-ENaC channels with higher efficacy than the ASIC1a channels, pointing on the heteromeric channels as a primary target of the toxin in cancer cells. Finally, bioinformatics analysis showed that the increased expression of ASIC1 and γ-ENaC correlates with a worse survival prognosis for patients with lung adenocarcinoma. Thus, the ASIC1a/α-ENaC/γ-ENaC heterotrimer can be considered a marker of cell oncogenicity and its targeting is promising for the design of new selective cancer therapeutics.

6.
Biochem Biophys Res Commun ; 619: 22-26, 2022 09 03.
Article in English | MEDLINE | ID: mdl-35728280

ABSTRACT

Tri-dimensional (3D) cell aggregates or spheroids are considered to be closer to physiological conditions than traditional 2D cell culture. Mesenchymal stem cells (MSCs) assembling in spheroids have increased the survival of transplanted cells. The organization of stem cells in 3D culture affects cell microenvironment and their mechanical properties. The regulation of the biological processes that maintain crucial physiological reactions of MSCs is closely related to the functioning of ion channels. The pattern of expression, role and regulatory mechanisms of ion channels could be significantly different in 3D compared to 2D culture, and, thus, needed to be properly analyzed on the level of ionic currents. Electrophysiological data on the features of ion channels functioning in 3D cell culture models are currently very limited in the literature. This gap of knowledge may be associated with technical difficulties that exist when researchers try to apply the standard patch clamp method for the registration of ion channels in cells aggregated in spheroids. In this regard, our study focuses on solving emerging technical difficulties and presents an example of their successful solution. Here, we developed a specific approach and have recorded the activity of mechanosensitive stretch-activated ion channels (SACs) in endometrial MSCs (eMSCs) assembled in spheroids. Moreover, we observed functional interplay of SACs with potassium channels of big conductance (BK) in the plasma membrane of eMSC spheroids consistently to revealed earlier in routine 2D cultured cells. Additionally, we observed a significant decrease in the frequency of SACs activation in spheroids that may indicate the differences in the level of functional expression of channels in 3D culture comparing to 2D culture of eMSCs.


Subject(s)
Ion Channels , Mesenchymal Stem Cells , Cells, Cultured , Female , Humans , Ion Channels/metabolism , Patch-Clamp Techniques , Stem Cells
7.
Int J Mol Sci ; 23(7)2022 Mar 29.
Article in English | MEDLINE | ID: mdl-35409116

ABSTRACT

Endometrial mesenchymal stem cells (eMSCs) are a specific class of stromal cells which have the capability to migrate, develop and differentiate into different types of cells such as adipocytes, osteocytes or chondrocytes. It is this unique plasticity that makes the eMSCs significant for cellular therapy and regenerative medicine. Stem cells choose their way of development by analyzing the extracellular and intracellular signals generated by a mechanical force from the microenvironment. Mechanosensitive channels are part of the cellular toolkit that feels the mechanical environment and can transduce mechanical stimuli to intracellular signaling pathways. Here, we identify previously recorded, mechanosensitive (MS), stretch-activated channels as Piezo1 proteins in the plasma membrane of eMSCs. Piezo1 activity triggered by the channel agonist Yoda1 elicits influx of Ca2+, a known modulator of cytoskeleton reorganization and cell motility. We found that store-operated Ca2+ entry (SOCE) formed by Ca2+-selective channel ORAI1 and Ca2+ sensors STIM1/STIM2 contributes to Piezo1-induced Ca2+ influx in eMSCs. Particularly, the Yoda1-induced increase in intracellular Ca2+ ([Ca2+]i) is partially abolished by 2-APB, a well-known inhibitor of SOCE. Flow cytometry analysis and wound healing assay showed that long-term activation of Piezo1 or SOCE does not have a cytotoxic effect on eMSCs but suppresses their migratory capacity and the rate of cell proliferation. We propose that the Piezo1 and SOCE are both important determinants in [Ca2+]i regulation, which critically affects the migratory activity of eMSCs and, therefore, could influence the regenerative potential of these cells.


Subject(s)
Calcium Signaling , Calcium , Calcium/metabolism , Calcium Signaling/physiology , Cell Membrane/metabolism , Humans , Ion Channels/metabolism , ORAI1 Protein/metabolism , Stem Cells/metabolism , Stromal Interaction Molecule 1/metabolism
8.
PLoS One ; 16(11): e0260727, 2021.
Article in English | MEDLINE | ID: mdl-34847207

ABSTRACT

The actin cytoskeleton is indispensable for the motility and migration of all types of cells; therefore, it plays a crucial role in the ability of the tissues to repair. Mesenchymal stem cells are intensively used in regenerative medicine, but usually relatively low percent of transplanted cells reaches the injury. To overcome this evident limitation, researchers try to enhance the motility and migration rate of the cells. As one of the approaches, co-cultivation and preconditioning of stem cells with biologically active compounds, which can cause actin cytoskeleton rearrangements followed by an increase of migratory properties of the cells, could be applied. The observed changes in F-actin structure induced by the compounds require quantitative estimation, and measurement of fluorescence intensity of the F-actin image captured by various microscopic techniques is commonly used nowadays. However, this approach could not always accurately detect the observed changes in the shape and structure of actin cytoskeleton. At this time, the image of F-actin has an irregular geometric pattern, and thus could be considered and characterized as a fractal object. To quantify the re-organization of cellular F-actin in terms of fractal geometry Minkovsky's box-counting method is suitable, but it is not widely used nowadays. We modified and improved the previously described method for fractal dimension measurement, and successfully applied it for the quantification of the F-actin structures of human mesenchymal stem cells.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Movement , Mesenchymal Stem Cells/metabolism , Child, Preschool , Humans , Male , Protein Conformation
9.
Int J Mol Sci ; 22(15)2021 Jul 22.
Article in English | MEDLINE | ID: mdl-34360605

ABSTRACT

Piezo1/2 are mechanosensitive calcium-permeable channels that can be activated by various modes of membrane deformation. The identification of the small molecule Yoda1, a synthetic Piezo1 agonist, revealed the possibility of chemical activation of the channel. Stimulating effects of Yoda1 on Piezo1 have been mainly documented using over-expressing cellular systems or channel proteins incorporated in artificial lipid bilayers. However, the activating effect of Yoda1 on native Piezo1 channels in the plasma membrane of living cells remains generally undefined, despite the increasing number of studies in which the agonist is utilized as a functional tool to reveal the contribution of Piezo1 to cellular reactions. In the current study, we used the human myeloid leukemia K562 cell line as a suitable model to examine chemically induced Piezo1 activity with the use of the patch-clamp technique in various specific modes. The functional expression of Piezo1 in leukemia cells was evidenced using a combinative approach, including single channel patch-clamp measurements. Utilizing our established single-current whole-cell assay on K562 cells, we have shown, for the first time, the selective real-time chemical activation of endogenously expressed Piezo1. Extracellular application of 0.5-1 µM Yoda1 effectively stimulated single Piezo1 currents in the cell membrane.


Subject(s)
Cell Membrane/metabolism , Ion Channels/drug effects , Leukemia/drug therapy , Mechanotransduction, Cellular , Pyrazines/pharmacology , Single-Cell Analysis/methods , Thiadiazoles/pharmacology , Cell Membrane/drug effects , Humans , Ion Channels/agonists , Ion Channels/metabolism , Leukemia/metabolism , Leukemia/pathology
11.
Biochem Biophys Res Commun ; 514(1): 173-179, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31029419

ABSTRACT

Increased migratory, invasive and metastatic potential is one of the main pathophysiological determinants of malignant cells. Mechanosensitive calcium-permeable ion channels are among the key membrane proteins that participate in processes of cellular motility. Local calcium influx via mechanosensitive channels was proposed to regulate calcium-dependent molecules involved in cell migration. Piezo transmembrane proteins were shown to act as calcium-permeable mechanosensitive ion channels in various cells and tissues, including a number of tumor cells. Furthermore, an elevated expression of Piezo1 is correlated with poor prognosis for some types of cancers. At the same time, functional impact of Piezo1 channels on pathophysiological reactions of tumor cells remains largely unknown. Here, we used 3T3B-SV40 mouse fibroblasts as a model to study the effect of Yoda1, selective Piezo1 activator, on migrative properties of transformed cells. RT-PCR and immunofluorescent staining showed the presence of native Piezo1 in 3T3B-SV40 fibroblasts. Functional expression of Piezo1 in plasma membrane of 3T3B-SV40 cells was confirmed by calcium measurements and single channel patch-clamp analysis. Particularly, application of Yoda1 resulted in rapid calcium influx and induced typical channel activity in membrane patches with characteristics identical to stretch-activated channels in 3T3B-SV40 cells. Importantly, dose-dependent inhibition of cellular migration by Yoda1 was found in wound healing assay using live cell imaging. Consistently, microscopic analysis showed that Yoda1 significantly altered cellular morphology, induced F-actin assembly and stress fiber formation indicating partial reversion of transformed phenotype. The results demonstrate for the first time that Piezo1 activation by selective agonist Yoda1 could be favorable for inhibiting migrative potential of transformed cells with native Piezo1 expression.


Subject(s)
Fibroblasts/drug effects , Fibroblasts/pathology , Ion Channels/metabolism , Pyrazines/pharmacology , Thiadiazoles/pharmacology , Animals , Calcium/metabolism , Cell Line, Transformed , Cell Movement/drug effects , Ion Channels/agonists , Ion Channels/genetics , Mice , Patch-Clamp Techniques
12.
Sci Rep ; 9(1): 4595, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872711

ABSTRACT

The study of ion channels in stem cells provides important information about their role in stem cell fate. Previously we have identified the activity of calcium-activated potassium channels of big conductance (BK channels) in human endometrium-derived mesenchymal stem cells (eMSCs). BK channels could have significant impact into signaling processes by modulating membrane potential. The membrane potential and ionic permeability dynamically changes during cycle transitions. Here, we aimed at verification of the role of BK channels as potassium transporting pathway regulating cell cycle passageway of eMSCs. The functional expression of native BK channels was confirmed by patch-clamp and immunocytochemistry. In non-synchronized cells immunofluorescent analysis revealed BK-positive and BK-negative stained eMSCs. Using cell synchronization, we found that the presence of BK channels in plasma membrane was cell cycle-dependent and significantly decreased in G2M phase. However, the study of cell cycle progression in presence of selective BK channel inhibitors showed no effect of pore blockers on cycle transitions. Thus, BK channel-mediated K+ transport is not critical for the fundamental mechanism of passageway through cell cycle of eMSCs. At the same time, the dynamics of the presence of BK channels on plasma membrane of eMSCs can be a novel indicator of cellular proliferation.


Subject(s)
Cell Cycle/genetics , Endometrium/cytology , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Electrophysiological Phenomena , Female , Humans , Large-Conductance Calcium-Activated Potassium Channels/metabolism
13.
J Cell Biochem ; 120(1): 461-469, 2019 01.
Article in English | MEDLINE | ID: mdl-30203535

ABSTRACT

Sodium influx is tightly regulated in the cells of blood origin. Amiloride-insensitive sodium channels were identified as one of the main sodium-transporting pathways in leukemia cells. To date, all known regulatory pathways of these channels are coupled with intracellular actin cytoskeleton dynamics. Here, to search for physiological mechanisms controlling epithelial Na+ channel (ENaC)-like channels, we utilized leukemia K562 cells as a unique model to examine single channel behavior in a whole-cell patch-clamp experiments. We have shown for the first time that extracellular serine protease trypsin directly activates sodium channels in plasma membrane of K562 cells. The whole-cell single current recordings clearly demonstrate no inhibition of trypsin-activated channels by amiloride or benzamil. Involvement of proteolytic cleavage in channel opening was confirmed in experiments with soybean trypsin inhibitor. More importantly, stabilization of F-actin with intracellular phalloidin did not prevent trypsin-induced channel activation indicating no implication of cytoskeleton rearrangements in stimulatory effect of extracellular protease. Our data reveals a novel mechanism modulating amiloride-insensitive ENaC-like channel activity and integral sodium permeability in leukemia cells.


Subject(s)
Amiloride/pharmacology , Epithelial Sodium Channels/metabolism , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Trypsin/pharmacology , Actin Cytoskeleton/metabolism , Actins/metabolism , Amiloride/analogs & derivatives , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cytochalasin D/pharmacology , Epithelial Sodium Channel Blockers/pharmacology , Humans , K562 Cells , Membrane Potentials/drug effects , Microscopy, Fluorescence , Models, Biological , Patch-Clamp Techniques , Phalloidine/pharmacology , Sodium/metabolism , Trypsin/metabolism , Trypsin Inhibitors/pharmacology
14.
Cell Biol Int ; 41(9): 1020-1029, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28656734

ABSTRACT

Statins are the most commonly prescribed agents used to modulate cholesterol levels in course of hypercholesterolemia treatment because of their relative tolerability and LDL-C lowering effect. Recently, there are emerging interests in the perspectives of statin drugs as anticancer agents based on preclinical evidence of their antiproliferative, proapoptotic, and anti-invasive properties. Functional impact of statin application on transformed cells still remains obscure that requires systematic study on adequate cellular models to provide correct comparison with their non-transformed counterparts. Cholesterol is the major lipid component of mammalian cells and it plays a crucial role in organization, lateral heterogeneity, and dynamics of plasma membrane as well as in membrane-cytoskeleton interrelations. To date, it is uncertain whether cellular effects of statins involve lipid-dependent alteration of plasma membrane. Here, the effects of simvastatin on lipid rafts, F-actin network and cellular viability were determined in comparative experiments on transformed fibroblasts and their non-transformed counterpart. GM1 lipid raft marker staining indicated no change of lipid raft integrity after short- or long-term simvastatin treatments. In the same time, simvastatin induced cytoskeleton rearrangement including partial F-actin disruption in cholesterol- and lipid raft-independent manner. Simvastatin dose-dependently affected viability of BALB/3T3 and 3T3B-SV40 cell lines: transformed fibroblasts were noticeably more sensitive to simvastatin comparing to non-transformed cells.


Subject(s)
Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Membrane Microdomains/drug effects , Simvastatin/pharmacology , Actins/metabolism , Animals , BALB 3T3 Cells , Cell Line , Cell Membrane/metabolism , Cell Survival/drug effects , Cholesterol/metabolism , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Membrane Microdomains/metabolism , Mice , Simian virus 40 , Transfection
15.
Biochem Biophys Res Commun ; 482(4): 563-568, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27856251

ABSTRACT

Mechanical forces are implicated in key physiological processes in stem cells, including proliferation, differentiation and lineage switching. To date, there is an evident lack of understanding of how external mechanical cues are coupled with calcium signalling in stem cells. Mechanical reactions are of particular interest in adult mesenchymal stem cells because of their promising potential for use in tissue remodelling and clinical therapy. Here, single channel patch-clamp technique was employed to search for cation channels involved in mechanosensitivity in mesenchymal endometrial-derived stem cells (hMESCs). Functional expression of native mechanosensitive stretch-activated channels (SACs) and calcium-sensitive potassium channels of different conductances in hMESCs was shown. Single current analysis of stretch-induced channel activity revealed functional coupling of SACs and BK channels in plasma membrane. The combination of cell-attached and inside-out experiments have indicated that highly localized Ca2+ entry via SACs triggers BK channel activity. At the same time, SK channels are not coupled with SACs despite of high calcium sensitivity as compared to BK. Our data demonstrate novel mechanism controlling BK channel activity in native cells. We conclude that SACs and BK channels are clusterized in functional mechanosensitive domains in the plasma membrane of hMESCs. Co-clustering of ion channels may significantly contribute to mechano-dependent calcium signalling in stem cells.


Subject(s)
Calcium Signaling , Ion Channels/metabolism , Mechanotransduction, Cellular , Mesenchymal Stem Cells/metabolism , Calcium/metabolism , Cells, Cultured , Endometrium/cytology , Female , Humans , Mesenchymal Stem Cells/cytology
16.
Biochem Biophys Res Commun ; 461(1): 54-8, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25858317

ABSTRACT

Sodium influx mediated by ion channels of plasma membrane underlies fundamental physiological processes in cells of blood origin. However, little is known about the single channel activity and regulatory mechanisms of sodium-specific channels in native cells. In the present work, we used different modes of patch clamp technique to examine ion channels involved in Na-transporting pathway in U937 human lymphoma cells. The activity of native non-voltage-gated sodium (NVGS) channels with unitary conductance of 10 pS was revealed in cell-attached, inside-out and whole-cell configurations. NVGS channel activity is directly controlled by submembranous actin cytoskeleton. Specifically, an activation of sodium channels in U937 cells in response to microfilament disassembly was demonstrated on single-channel and integral current level. Inside-out experiments showed that filament assembly on cytoplasmic membrane surface caused fast inactivation of the channels. Biophysical characteristics of NVGS channels in U937 cells were similar to that of epithelial sodium channels (ENaCs). However, we found that amiloride, a known inhibitor of DEG/ENaC, did not block NVGS channels in U937 cells. Whole-cell current measurements revealed no amiloride-sensitive component of membrane current. Our data show that cortical actin structures represent the main factor that controls the activity of amiloride-insensitive ENaC-like channels in human lymphoma cells.


Subject(s)
Actin Cytoskeleton/metabolism , Amiloride/administration & dosage , Ion Channel Gating/drug effects , Lymphoma/metabolism , Sodium Channels/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Epithelial Sodium Channel Blockers/administration & dosage , Humans , Sodium/metabolism
17.
Biochem Biophys Res Commun ; 451(3): 421-4, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25108157

ABSTRACT

The major players in the processes of cellular mechanotransduction are considered to be mechanosensitive (MS) or mechano-gated ion channels. Non-selective Ca(2+)-permeable channels, whose activity is directly controlled by membrane stretch (stretch-activated channels, SACs) are ubiquitously present in mammalian cells of different origin. Ca(2+) entry mediated by SACs presumably has a significant impact on various Ca(2+)-dependent intracellular and membrane processes. It was proposed that SACs could play a crucial role in the different cellular reactions and pathologies, including oncotransformation, increased metastatic activity and invasion of malignant cells. In the present work, coupling of ion channels in transformed fibroblasts in course of stretch activation was explored with the use of patch-clamp technique. The combination of cell-attached and inside-out single-current experiments showed that Ca(2+) influx via SACs triggered the activity of Ca(2+)-sensitive K(+) channels indicating functional compartmentalization of different channel types in plasma membrane. Importantly, the analysis of single channel behavior demonstrated that K(+) currents could be activated by the rise of intracellular calcium but displayed no direct mechanosensitivity. Taken together, our data imply that local changes in Ca(2+) concentration due to SAC activity may provide a functional link between various Ca(2+)-dependent molecules in the processes of cellular mechanotransduction.


Subject(s)
Calcium Channels/physiology , Ion Channels/physiology , Mechanotransduction, Cellular/physiology , Potassium Channels, Calcium-Activated/physiology , Animals , Fibroblasts/physiology , Ion Channel Gating , Mice , Patch-Clamp Techniques
18.
Kidney Int ; 86(3): 506-14, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24646854

ABSTRACT

A key role for podocytes in the pathogenesis of proteinuric renal diseases has been established. Angiotensin II causes depolarization and increased intracellular calcium concentration in podocytes; members of the cation TRPC channels family, particularly TRPC6, are proposed as proteins responsible for calcium flux. Angiotensin II evokes calcium transient through TRPC channels and mutations in the gene encoding the TRPC6 channel result in the development of focal segmental glomerulosclerosis. Here we examined the effects of angiotensin II on intracellular calcium ion levels and endogenous channels in intact podocytes of freshly isolated decapsulated mouse glomeruli. An ion channel with distinct TRPC6 properties was identified in wild-type, but was absent in TRPC6 knockout mice. Single-channel electrophysiological analysis found that angiotensin II acutely activated native TRPC-like channels in both podocytes of freshly isolated glomeruli and TRPC6 channels transiently overexpressed in CHO cells; the effect was mediated by changes in the channel open probability. Angiotensin II evoked intracellular calcium transients in the wild-type podocytes, which was blunted in TRPC6 knockout glomeruli. Pan-TRPC inhibitors gadolinium and SKF 96365 reduced the response in wild-type glomerular epithelial cells, whereas the transient in TRPC6 knockout animals was not affected. Thus, angiotensin II-dependent activation of TRPC6 channels in podocytes may have a significant role in the development of kidney diseases.


Subject(s)
Angiotensin II/pharmacology , Calcium/metabolism , Podocytes/drug effects , Podocytes/metabolism , TRPC Cation Channels/drug effects , TRPC Cation Channels/metabolism , Animals , CHO Cells , Calcium Channel Blockers/pharmacology , Cricetulus , Gadolinium/pharmacology , Imidazoles/pharmacology , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Angiotensin, Type 1/metabolism , TRPC Cation Channels/genetics , TRPC6 Cation Channel , Up-Regulation/drug effects
19.
Cell Tissue Res ; 354(3): 783-92, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24036843

ABSTRACT

Dynamic remodeling of the actin cytoskeleton plays an essential role in cell migration and various signaling processes in living cells. One of the critical factors that controls the nucleation of new actin filaments in eukaryotic cells is the actin-related protein 2/3 (Arp2/3) complex. Recently, two novel classes of small molecules that bind to different sites on the Arp2/3 complex and inhibit its ability to nucleate F-actin have been discovered and described. The current study aims at investigating the effects of CK-0944666 (CK-666) and its analogs (CK-869 and inactive CK-689) on the reorganization of the actin microfilaments in the cortical collecting duct cell line, M-1. We show that treatment with CK-666 and CK869 results in the reorganization of F-actin and drastically affects cell motility rate. The concentrations of the compounds used in this study (100-200 µM) neither cause loss of cell viability nor influence cell shape or monolayer integrity; hence, the effects of described compounds were not due to structural side effects. Therefore, we conclude that the Arp2/3 complex plays an important role in cell motility and F-actin reorganization in M-1 cells. Furthermore, CK-666 and its analogs are useful tools for the investigation of the Arp2/3 complex.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Kidney Tubules, Collecting/metabolism , Animals , Cell Movement/physiology , Immunohistochemistry , Indoles/metabolism , Kidney Tubules, Collecting/cytology , Mice , Signal Transduction
20.
Cell Biol Int ; 37(6): 617-23, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23447521

ABSTRACT

Membrane cholesterol and lipid rafts are implicated in various signalling processes involving actin rearrangement in living cells. However, functional link between raft integrity and organisation of cytoskeleton remains unclear. We have compared the effect of cholesterol sequestration on F-actin structures in normal and transformed fibroblasts in which microfilament system is developed to a different extent. The depletion of membrane cholesterol by methyl-beta-cyclodextrin (MbCD) resulted in a disruption of lipid rafts in plasma membrane as it was revealed by fluorescent labelling of GM1 ganglioside. In normal fibroblasts with highly developed microfilament system, the cholesterol depletion resulted in actin disassembly and reduction of stress fibres. However, in transformed cells containing low amount of fibrillar actin, MbCD treatment induced intensive formation of stress fibres and increased cell spreading. The results show that the effect of cholesterol depletion and lipid raft disruption on microfilament system is critically determined by the initial state of cytoskeleton, specifically, by the balance of polymerised and monomeric actin in the cell. We assume that uncapping of the microfilaments is the key step of cholesterol-regulated actin remodelling.


Subject(s)
Actin Cytoskeleton/metabolism , Cholesterol/metabolism , Fibroblasts/metabolism , Animals , BALB 3T3 Cells , Cell Line, Transformed , Fibroblasts/cytology , Mice , beta-Cyclodextrins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...