51
|
Sequential breakdown of 3-phosphorylated phosphoinositides is essential for the completion of macropinocytosis. Proc Natl Acad Sci U S A 2014; 111:E978-87. [PMID: 24591580 DOI: 10.1073/pnas.1311029111] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Macropinocytosis is a highly conserved endocytic process by which extracellular fluid and solutes are internalized into cells. Macropinocytosis starts with the formation of membrane ruffles at the plasma membrane and ends with their closure. The transient and sequential emergence of phosphoinositides PI(3,4,5)P3 and PI(3,4)P2 in the membrane ruffles is essential for macropinocytosis. By making use of information in the Caenorhabditis elegans mutants defective in fluid-phase endocytosis, we found that mammalian phosphoinositide phosphatase MTMR6 that dephosphorylates PI(3)P to PI, and its binding partner MTMR9, are required for macropinocytosis. INPP4B, which dephosphorylates PI(3,4)P2 to PI(3)P, was also found to be essential for macropinocytosis. These phosphatases operate after the formation of membrane ruffles to complete macropinocytosis. Finally, we showed that KCa3.1, a Ca(2+)-activated K(+) channel that is activated by PI(3)P, is required for macropinocytosis. We propose that the sequential breakdown of PI(3,4,5)P3 → PI(3,4)P2 → PI(3)P → PI controls macropinocytosis through specific effectors of the intermediate phosphoinositides.
Collapse
|
52
|
Turner KL, Honasoge A, Robert SM, McFerrin MM, Sontheimer H. A proinvasive role for the Ca(2+) -activated K(+) channel KCa3.1 in malignant glioma. Glia 2014; 62:971-81. [PMID: 24585442 DOI: 10.1002/glia.22655] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/03/2014] [Accepted: 02/14/2014] [Indexed: 11/09/2022]
Abstract
Glioblastoma multiforme are highly motile primary brain tumors. Diffuse tissue invasion hampers surgical resection leading to poor patient prognosis. Recent studies suggest that intracellular Ca(2+) acts as a master regulator for cell motility and engages a number of downstream signals including Ca(2+) -activated ion channels. Querying the REepository of Molecular BRAin Neoplasia DaTa (REMBRANDT), an annotated patient gene database maintained by the National Cancer Institute, we identified the intermediate conductance Ca(2+) -activated K(+) channels, KCa3.1, being overexpressed in 32% of glioma patients where protein expression significantly correlated with poor patient survival. To mechanistically link KCa3.1 expression to glioma invasion, we selected patient gliomas that, when propagated as xenolines in vivo, present with either high or low KCa3.1 expression. In addition, we generated U251 glioma cells that stably express an inducible knockdown shRNA to experimentally eliminate KCa3.1 expression. Subjecting these cells to a combination of in vitro and in situ invasion assays, we demonstrate that KCa3.1 expression significantly enhances glioma invasion and that either specific pharmacological inhibition with TRAM-34 or elimination of the channel impairs invasion. Importantly, after intracranial implantation into SCID mice, ablation of KCa3.1 with inducible shRNA resulted in a significant reduction in tumor invasion into surrounding brain in vivo. These results show that KCa3.1 confers an invasive phenotype that significantly worsens a patient's outlook, and suggests that KCa3.1 represents a viable therapeutic target to reduce glioma invasion.
Collapse
Affiliation(s)
- Kathryn L Turner
- Department of Neurobiology and Center for Glial Biology in Medicine, University of Alabama at Birmingham, Alabama
| | | | | | | | | |
Collapse
|
53
|
Arcangeli A, Crociani O, Bencini L. Interaction of tumour cells with their microenvironment: ion channels and cell adhesion molecules. A focus on pancreatic cancer. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130101. [PMID: 24493749 DOI: 10.1098/rstb.2013.0101] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cancer must be viewed as a 'tissue', constituted of both transformed cells and a heterogeneous microenvironment, the 'tumour microenvironment' (TME). The TME undergoes a complex remodelling during the course of multistep tumourigenesis, hence strongly contributing to tumour progression. Ion channels and transporters (ICTs), being expressed on both tumour cells and in the different cellular components of the TME, are in a strategic position to sense and mediate signals arising from the TME. Often, this transmission is mediated by integrin adhesion receptors, which are the main cellular receptors capable of mediating cell-to-cell and cell-to-matrix bidirectional signalling. Integrins can often operate in conjunction with ICT because they can behave as functional partners of ICT proteins. The role of integrin receptors in the crosstalk between tumour cells and the TME is particularly relevant in the context of pancreatic cancer (PC), characterized by an overwhelming TME which actively contributes to therapy resistance. We discuss the possibility that this occurs through integrins and ICTs, which could be exploited as targets to overcome chemoresistance in PC.
Collapse
Affiliation(s)
- Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, University of Florence, , Viale G.B. Morgagni, 50, 50134 Firenze, Italy
| | | | | |
Collapse
|
54
|
Dagley LF, Croft NP, Isserlin R, Olsen JB, Fong V, Emili A, Purcell AW. Discovery of novel disease-specific and membrane-associated candidate markers in a mouse model of multiple sclerosis. Mol Cell Proteomics 2013; 13:679-700. [PMID: 24361864 DOI: 10.1074/mcp.m113.033340] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Multiple sclerosis is a chronic demyelinating disorder characterized by the infiltration of auto-reactive immune cells from the periphery into the central nervous system resulting in axonal injury and neuronal cell death. Experimental autoimmune encephalomyelitis represents the best characterized animal model as common clinical, histological, and immunological features are recapitulated. A label-free mass spectrometric proteomics approach was used to detect differences in protein abundance within specific fractions of disease-affected tissues including the soluble lysate derived from the spinal cord and membrane protein-enriched peripheral blood mononuclear cells. Tissues were harvested from actively induced experimental autoimmune encephalomyelitis mice and sham-induced ("vehicle" control) counterparts at the disease peak followed by subsequent analysis by nanoflow liquid chromatography tandem mass spectrometry. Relative protein quantitation was performed using both intensity- and fragmentation-based approaches. After statistical evaluation of the data, over 500 and 250 differentially abundant proteins were identified in the spinal cord and peripheral blood mononuclear cell data sets, respectively. More than half of these observations have not previously been linked to the disease. The biological significance of all candidate disease markers has been elucidated through rigorous literature searches, pathway analysis, and validation studies. Results from comprehensive targeted mass spectrometry analyses have confirmed the differential abundance of ∼ 200 candidate markers (≥ twofold dysregulated expression) at a 70% success rate. This study is, to our knowledge, the first to examine the cell-surface proteome of peripheral blood mononuclear cells in experimental autoimmune encephalomyelitis. These data provide a unique mechanistic insight into the dynamics of peripheral immune cell infiltration into CNS-privileged sites at a molecular level and has identified several candidate markers, which represent promising targets for future multiple sclerosis therapies. The mass spectrometry proteomics data associated with this manuscript have been deposited to the ProteomeXchange Consortium with the data set identifier PXD000255.
Collapse
Affiliation(s)
- Laura F Dagley
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | | | | | | | | | | |
Collapse
|
55
|
Stock C, Ludwig FT, Hanley PJ, Schwab A. Roles of ion transport in control of cell motility. Compr Physiol 2013; 3:59-119. [PMID: 23720281 DOI: 10.1002/cphy.c110056] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell motility is an essential feature of life. It is essential for reproduction, propagation, embryonic development, and healing processes such as wound closure and a successful immune defense. If out of control, cell motility can become life-threatening as, for example, in metastasis or autoimmune diseases. Regardless of whether ciliary/flagellar or amoeboid movement, controlled motility always requires a concerted action of ion channels and transporters, cytoskeletal elements, and signaling cascades. Ion transport across the plasma membrane contributes to cell motility by affecting the membrane potential and voltage-sensitive ion channels, by inducing local volume changes with the help of aquaporins and by modulating cytosolic Ca(2+) and H(+) concentrations. Voltage-sensitive ion channels serve as voltage detectors in electric fields thus enabling galvanotaxis; local swelling facilitates the outgrowth of protrusions at the leading edge while local shrinkage accompanies the retraction of the cell rear; the cytosolic Ca(2+) concentration exerts its main effect on cytoskeletal dynamics via motor proteins such as myosin or dynein; and both, the intracellular and the extracellular H(+) concentration modulate cell migration and adhesion by tuning the activity of enzymes and signaling molecules in the cytosol as well as the activation state of adhesion molecules at the cell surface. In addition to the actual process of ion transport, both, channels and transporters contribute to cell migration by being part of focal adhesion complexes and/or physically interacting with components of the cytoskeleton. The present article provides an overview of how the numerous ion-transport mechanisms contribute to the various modes of cell motility.
Collapse
Affiliation(s)
- Christian Stock
- Institute of Physiology II, University of Münster, Münster, Germany.
| | | | | | | |
Collapse
|
56
|
Girault A, Brochiero E. Evidence of K+ channel function in epithelial cell migration, proliferation, and repair. Am J Physiol Cell Physiol 2013; 306:C307-19. [PMID: 24196531 DOI: 10.1152/ajpcell.00226.2013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Efficient repair of epithelial tissue, which is frequently exposed to insults, is necessary to maintain its functional integrity. It is therefore necessary to better understand the biological and molecular determinants of tissue regeneration and to develop new strategies to promote epithelial repair. Interestingly, a growing body of evidence indicates that many members of the large and widely expressed family of K(+) channels are involved in regulation of cell migration and proliferation, key processes of epithelial repair. First, we briefly summarize the complex mechanisms, including cell migration, proliferation, and differentiation, engaged after epithelial injury. We then present evidence implicating K(+) channels in the regulation of these key repair processes. We also describe the mechanisms whereby K(+) channels may control epithelial repair processes. In particular, changes in membrane potential, K(+) concentration, cell volume, intracellular Ca(2+), and signaling pathways following modulation of K(+) channel activity, as well as physical interaction of K(+) channels with the cytoskeleton or integrins are presented. Finally, we discuss the challenges to efficient, specific, and safe targeting of K(+) channels for therapeutic applications to improve epithelial repair in vivo.
Collapse
Affiliation(s)
- Alban Girault
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; and
| | | |
Collapse
|
57
|
Hypotonic Regulation of Mouse Epithelial Sodium Channel in Xenopus laevis Oocytes. J Membr Biol 2013; 246:949-58. [DOI: 10.1007/s00232-013-9598-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/24/2013] [Indexed: 10/26/2022]
|
58
|
Huber SM, Butz L, Stegen B, Klumpp D, Braun N, Ruth P, Eckert F. Ionizing radiation, ion transports, and radioresistance of cancer cells. Front Physiol 2013; 4:212. [PMID: 23966948 PMCID: PMC3743404 DOI: 10.3389/fphys.2013.00212] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/24/2013] [Indexed: 12/22/2022] Open
Abstract
The standard treatment of many tumor entities comprises fractionated radiation therapy which applies ionizing radiation to the tumor-bearing target volume. Ionizing radiation causes double-strand breaks in the DNA backbone that result in cell death if the number of DNA double-strand breaks exceeds the DNA repair capacity of the tumor cell. Ionizing radiation reportedly does not only act on the DNA in the nucleus but also on the plasma membrane. In particular, ionizing radiation-induced modifications of ion channels and transporters have been reported. Importantly, these altered transports seem to contribute to the survival of the irradiated tumor cells. The present review article summarizes our current knowledge on the underlying mechanisms and introduces strategies to radiosensitize tumor cells by targeting plasma membrane ion transports.
Collapse
Affiliation(s)
- Stephan M Huber
- Department of Radiation Oncology, University of Tübingen Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
59
|
Yang M, Brackenbury WJ. Membrane potential and cancer progression. Front Physiol 2013; 4:185. [PMID: 23882223 PMCID: PMC3713347 DOI: 10.3389/fphys.2013.00185] [Citation(s) in RCA: 396] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 06/28/2013] [Indexed: 12/27/2022] Open
Abstract
Membrane potential (Vm), the voltage across the plasma membrane, arises because of the presence of different ion channels/transporters with specific ion selectivity and permeability. Vm is a key biophysical signal in non-excitable cells, modulating important cellular activities, such as proliferation and differentiation. Therefore, the multiplicities of various ion channels/transporters expressed on different cells are finely tuned in order to regulate the Vm. It is well-established that cancer cells possess distinct bioelectrical properties. Notably, electrophysiological analyses in many cancer cell types have revealed a depolarized Vm that favors cell proliferation. Ion channels/transporters control cell volume and migration, and emerging data also suggest that the level of Vm has functional roles in cancer cell migration. In addition, hyperpolarization is necessary for stem cell differentiation. For example, both osteogenesis and adipogenesis are hindered in human mesenchymal stem cells (hMSCs) under depolarizing conditions. Therefore, in the context of cancer, membrane depolarization might be important for the emergence and maintenance of cancer stem cells (CSCs), giving rise to sustained tumor growth. This review aims to provide a broad understanding of the Vm as a bioelectrical signal in cancer cells by examining several key types of ion channels that contribute to its regulation. The mechanisms by which Vm regulates cancer cell proliferation, migration, and differentiation will be discussed. In the long term, Vm might be a valuable clinical marker for tumor detection with prognostic value, and could even be artificially modified in order to inhibit tumor growth and metastasis.
Collapse
Affiliation(s)
- Ming Yang
- Department of Biology, University of York York, UK
| | | |
Collapse
|
60
|
Jacobsen KS, Zeeberg K, Sauter DRP, Poulsen KA, Hoffmann EK, Schwab A. The role of TMEM16A (ANO1) and TMEM16F (ANO6) in cell migration. Pflugers Arch 2013; 465:1753-62. [PMID: 23832500 PMCID: PMC3898376 DOI: 10.1007/s00424-013-1315-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/14/2013] [Accepted: 06/15/2013] [Indexed: 12/30/2022]
Abstract
Members of the TMEM16 family have recently been described as Ca2+-activated Cl− channels. They have been implicated in cancer and appear to be associated with poor patient prognosis. Here, we investigate the role of TMEM16 channels in cell migration, which is largely unknown. We focused on TMEM16A and TMEM16F channels that have the highest expression of TMEM16 channels in Ehrlich Lettre ascites (ELA) cells. Due to the lack of specific pharmacological modulators, we employed a miRNA approach and stably knocked down the expression of TMEM16A and TMEM16F channels, respectively. Migration analysis shows that TMEM16A KD clones are affected in their directional migration, whereas TMEM16F KD clones show a 40 % reduced rate of cell migration. Moreover, TMEM16A KD clones have a smaller projected cell area, and they are rounder than TMEM16F KD clones. The morphological changes are linearly correlated with the directionality of cells. TMEM16A and TMEM16F, thus, have an important function in cell migration—TMEM16A in directional migration, TMEM16F in determination of the speed of migration. We conclude that TMEM16A and TMEM16F channels have a distinct impact on the steering and motor mechanisms of migrating ELA cells.
Collapse
Affiliation(s)
- K S Jacobsen
- Department of Biology, August Krogh Building, Universitetsparken 13, 2100, Copenhagen Ø, Denmark
| | | | | | | | | | | |
Collapse
|
61
|
Schäffer TE. Nanomechanics of molecules and living cells with scanning ion conductance microscopy. Anal Chem 2013; 85:6988-94. [PMID: 23692368 DOI: 10.1021/ac400686k] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hydrodynamic flow through a nanopipet in a scanning ion conductance microscope (SICM) can exert localized forces on a sample surface. These forces can be used for trapping of molecules in lipid bilayers and for mapping the mechanical properties of living cells.
Collapse
Affiliation(s)
- Tilman E Schäffer
- University of Tübingen, Department of Physics and LISA+, Tübingen, Germany
| |
Collapse
|
62
|
ENaC contribution to epithelial wound healing is independent of the healing mode and of any increased expression in the channel. Cell Tissue Res 2013; 353:53-64. [DOI: 10.1007/s00441-013-1635-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 04/05/2013] [Indexed: 12/13/2022]
|
63
|
Turner KL, Sontheimer H. KCa3.1 modulates neuroblast migration along the rostral migratory stream (RMS) in vivo. ACTA ACUST UNITED AC 2013; 24:2388-400. [PMID: 23585521 DOI: 10.1093/cercor/bht090] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
From the subventricular zone (SVZ), neuronal precursor cells (NPCs), called neuroblasts, migrate through the rostral migratory stream (RMS) to become interneurons in the olfactory bulb (OB). Ion channels regulate neuronal migration during development, yet their role in migration through the adult RMS is unknown. To address this question, we utilized Nestin-CreER(T2)/R26R-YFP mice to fluorescently label neuroblasts in the adult. Patch-clamp recordings from neuroblasts reveal K(+) currents that are sensitive to intracellular Ca(2+) levels and blocked by clotrimazole and TRAM-34, inhibitors of intermediate conductance Ca(2+)-activated K(+) (KCa3.1) channels. Immunolabeling and electrophysiology show KCa3.1 expression restricted to neuroblasts in the SVZ and RMS, but absent in OB neurons. Time-lapse confocal microscopy in situ showed inhibiting KCa3.1 prolonged the stationary phase of neuroblasts' saltatory migration, reducing migration speed by over 50%. Both migration and KCa3.1 currents could also be inhibited by blocking Ca(2+) influx via transient receptor potential (TRP) channels, which, together with positive immunostaining for transient receptor potential canonical 1 (TRPC1), suggest that TRP channels are an important Ca(2+) source modulating KCa3.1 activity. Finally, injecting TRAM-34 into Nestin-CreER(T2)/R26R-YFP mice significantly reduced the number of neuroblasts that reached the OB, suggesting an important role for KCa3.1 in vivo. These studies describe a previously unrecognized protein in migration of adult NPCs.
Collapse
Affiliation(s)
- Kathryn L Turner
- Department of Neurobiology and Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Harald Sontheimer
- Department of Neurobiology and Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
64
|
|
65
|
Bradykinin-induced chemotaxis of human gliomas requires the activation of KCa3.1 and ClC-3. J Neurosci 2013; 33:1427-40. [PMID: 23345219 DOI: 10.1523/jneurosci.3980-12.2013] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Previous reports demonstrate that cell migration in the nervous system is associated with stereotypic changes in intracellular calcium concentration ([Ca(2+)](i)), yet the target of these changes are essentially unknown. We examined chemotactic migration/invasion of human gliomas to study how [Ca(2+)](i) regulates cellular movement and to identify downstream targets. Gliomas are primary brain cancers that spread exclusively within the brain, frequently migrating along blood vessels to which they are chemotactically attracted by bradykinin. Using simultaneous fura-2 Ca(2+) imaging and amphotericin B perforated patch-clamp electrophysiology, we find that bradykinin raises [Ca(2+)](i) and induces a biphasic voltage response. This voltage response is mediated by the coordinated activation of Ca(2+)-dependent, TRAM-34-sensitive K(Ca)3.1 channels, and Ca(2+)-dependent, 4,4'-diisothiocyanato-stilbene-2,2'-disulfonic acid (DIDS)-sensitive and gluconate-sensitive Cl(-) channels. A significant portion of these Cl(-) currents can be attributed to Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activation of ClC-3, a voltage-gated Cl(-) channel/transporter, because pharmacological inhibition of CaMKII or shRNA-mediated knockdown of ClC-3 inhibited Ca(2+)-activated Cl(-) currents. Western blots show that K(Ca)3.1 and ClC-3 are expressed in tissue samples obtained from patients diagnosed with grade IV gliomas. Both K(Ca)3.1 and ClC-3 colocalize to the invading processes of glioma cells. Importantly, inhibition of either channel abrogates bradykinin-induced chemotaxis and reduces tumor expansion in mouse brain slices in situ. These channels should be further explored as future targets for anti-invasive drugs. Furthermore, these data elucidate a novel mechanism placing cation and anion channels downstream of ligand-mediated [Ca(2+)](i) increases, which likely play similar roles in other migratory cells in the nervous system.
Collapse
|
66
|
Happel P, Möller K, Schwering NK, Dietzel ID. Migrating oligodendrocyte progenitor cells swell prior to soma dislocation. Sci Rep 2013; 3:1806. [PMID: 23657670 PMCID: PMC3648797 DOI: 10.1038/srep01806] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/24/2013] [Indexed: 11/09/2022] Open
Abstract
The migration of oligodendrocyte progenitor cells (OPCs) to the white matter is an indispensable requirement for an intact brain function. The mechanism of cell migration in general is not yet completely understood. Nevertheless, evidence is accumulating that besides the coordinated rearrangement of the cytoskeleton, a finetuned interplay of ion and water fluxes across the cell membrane is essential for cell migration. One part of a general hypothesis is that a local volume increase towards the direction of movement triggers a mechano-activated calcium influx that regulates various procedures at the rear end of a migrating cell. Here, we investigated cell volume changes of migrating OPCs using scanning ion conductance microscopy. We found that during accelerated migration OPCs undergo an increase in the frontal cell body volume. These findings are supplemented with time lapse calcium imaging data that hint an increase in calcium content the frontal part of the cell soma.
Collapse
Affiliation(s)
- Patrick Happel
- Central Unit for Ionbeams and Radionuclides (RUBION), Ruhr-University Bochum, Bochum, Germany.
| | | | | | | |
Collapse
|
67
|
Ding F, Zhang G, Liu L, Jiang L, Wang R, Zheng Y, Wang G, Xie M, Duan Y. Involvement of cationic channels in proliferation and migration of human mesenchymal stem cells. Tissue Cell 2012; 44:358-64. [DOI: 10.1016/j.tice.2012.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 06/07/2012] [Accepted: 06/07/2012] [Indexed: 10/28/2022]
|
68
|
Roubinet C, Tran PT, Piel M. Common mechanisms regulating cell cortex properties during cell division and cell migration. Cytoskeleton (Hoboken) 2012; 69:957-72. [PMID: 23125194 DOI: 10.1002/cm.21086] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 09/28/2012] [Accepted: 10/02/2012] [Indexed: 12/14/2022]
Abstract
Single cell morphogenesis results from a balance of forces involving internal pressure (also called turgor pressure in plants and fungi) and the plastic and dynamic outer shell of the cell. Dominated by the cell wall in plants and fungi, mechanical properties of the outer shell of animal cells arise from the cell cortex, which is mostly composed of the plasma membrane (and membrane proteins) and the underlying meshwork of actin filaments and myosin motors (and associated proteins). In this review, following Bray and White [1988; Science 239:883-889], we draw a parallel between the regulation of the cell cortex during cell division and cell migration in animal cells. Starting from the similarities in shape changes and underlying mechanical properties, we further propose that the analogy between cell division and cell migration might run deeper, down to the basic molecular mechanisms driving cell cortex remodeling. We focus our attention on how an heterogeneous and dynamic cortex can be generated to allow cell shape changes while preserving cell integrity.
Collapse
Affiliation(s)
- Chantal Roubinet
- Université de Toulouse, UPS, Centre de Biologie du Développement, Bâtiment 4R3, 118 route de Narbonne, F-31062 Toulouse, France.
| | | | | |
Collapse
|
69
|
Transient receptor potential canonical channel 1 impacts on mechanosignaling during cell migration. Pflugers Arch 2012; 464:623-30. [DOI: 10.1007/s00424-012-1169-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 09/26/2012] [Accepted: 09/27/2012] [Indexed: 01/01/2023]
|
70
|
Ruiz C, Martins JR, Rudin F, Schneider S, Dietsche T, Fischer CA, Tornillo L, Terracciano LM, Schreiber R, Bubendorf L, Kunzelmann K. Enhanced expression of ANO1 in head and neck squamous cell carcinoma causes cell migration and correlates with poor prognosis. PLoS One 2012; 7:e43265. [PMID: 22912841 PMCID: PMC3422276 DOI: 10.1371/journal.pone.0043265] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/18/2012] [Indexed: 12/23/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) has the potential for early metastasis and is associated with poor survival. Ano1 (Dog1) is an established and sensitive marker for the diagnosis of gastrointestinal stromal tumors (GIST) and has recently been identified as a Ca2+ activated Cl− channel. Although the ANO1 gene is located on the 11q13 locus, a region which is known to be amplified in different types of human carcinomas, a detailed analysis of Ano1 amplification and expression in HNSCC has not been performed. It is thus still unclear how Ano1 contributes to malignancy in HNSCC. We analyzed genomic amplification of the 11q13 locus and Ano1 together with Ano1-protein expression in a large collection of HNSCC samples. We detected a highly significant correlation between amplification and expression of Ano1 and showed that HNSCC patients with Ano1 protein expression have a poor overall survival. We further analyzed the expression of the Ano1 protein in more than 4′000 human samples from 80 different tumor types and 76 normal tissue types and detected that besides HNSCC and GISTs, Ano1 was rarely expressed in other tumor samples or healthy human tissues. In HNSCC cell lines, expression of Ano1 caused Ca2+ activated Cl− currents, which induced cell motility and cell migration in wound healing and in real time migration assays, respectively. In contrast, knockdown of Ano1 did not affect intracellular Ca2+ signaling and surprisingly did not reduce cell proliferation in BHY cells. Further, expression and activity of Ano1 strongly correlated with the ability of HNSCC cells to regulate their volume. Thus, poor survival in HNSCC patients is correlated with the presence of Ano1. Our results further suggest that Ano1 facilitates regulation of the cell volume and causes cell migration, which both can contribute to metastatic progression in HNSCC.
Collapse
Affiliation(s)
- Christian Ruiz
- Institute for Pathology, University Hospital Basel, Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Chen Y, Rice W, Gu Z, Li J, Huang J, Brenner MB, Van Hoek A, Xiong J, Gundersen GG, Norman JC, Hsu VW, Fenton RA, Brown D, Lu HAJ. Aquaporin 2 promotes cell migration and epithelial morphogenesis. J Am Soc Nephrol 2012; 23:1506-17. [PMID: 22859853 DOI: 10.1681/asn.2012010079] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The aquaporin 2 (AQP2) water channel, expressed in kidney collecting ducts, contributes critically to water homeostasis in mammals. Animals lacking or having significantly reduced levels of AQP2, however, have not only urinary concentrating abnormalities but also renal tubular defects that lead to neonatal mortality from renal failure. Here, we show that AQP2 is not only a water channel but also an integrin-binding membrane protein that promotes cell migration and epithelial morphogenesis. AQP2 expression modulates the trafficking and internalization of integrin β1, facilitating its turnover at focal adhesions. In vitro, disturbing the interaction between AQP2 and integrin β1 by mutating the RGD motif led to reduced endocytosis, retention of integrin β1 at the cell surface, and defective cell migration and tubulogenesis. Similarly, in vivo, AQP2-null mice exhibited significant retention of integrin β1 at the basolateral membrane and had tubular abnormalities. In summary, these data suggest that the water channel AQP2 interacts with integrins to promote renal epithelial cell migration, contributing to the structural and functional integrity of the mammalian kidney.
Collapse
Affiliation(s)
- Ying Chen
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Ben Soussia I, Mies F, Naeije R, Shlyonsky V. Melatonin down-regulates volume-sensitive chloride channels in fibroblasts. Pflugers Arch 2012; 464:273-85. [DOI: 10.1007/s00424-012-1139-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 01/01/2023]
|
73
|
Expression and Role of the Intermediate-Conductance Calcium-Activated Potassium Channel KCa3.1 in Glioblastoma. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:421564. [PMID: 22675627 PMCID: PMC3362965 DOI: 10.1155/2012/421564] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 03/15/2012] [Indexed: 12/29/2022]
Abstract
Glioblastomas are characterized by altered expression of several ion channels that have important consequences in cell functions associated with their aggressiveness, such as cell survival, proliferation, and migration. Data on the altered expression and function of the intermediate-conductance calcium-activated K (KCa3.1) channels in glioblastoma cells have only recently become available. This paper aims to (i) illustrate the main structural, biophysical, pharmacological, and modulatory properties of the KCa3.1 channel, (ii) provide a detailed account of data on the expression of this channel in glioblastoma cells, as compared to normal brain tissue, and (iii) critically discuss its major functional roles. Available data suggest that KCa3.1 channels (i) are highly expressed in glioblastoma cells but only scantly in the normal brain parenchima, (ii) play an important role in the control of glioblastoma cell migration. Altogether, these data suggest KCa3.1 channels as potential candidates for a targeted therapy against this tumor.
Collapse
|
74
|
Dilly S, Poncin S, Lamy C, Liégeois JF, Seutin V. Physiologie, pharmacologie et modélisation de canaux potassiques. Med Sci (Paris) 2012; 28:395-402. [DOI: 10.1051/medsci/2012284017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
75
|
Markova O, Lenne PF. Calcium signaling in developing embryos: focus on the regulation of cell shape changes and collective movements. Semin Cell Dev Biol 2012; 23:298-307. [PMID: 22414534 DOI: 10.1016/j.semcdb.2012.03.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 01/31/2012] [Accepted: 03/04/2012] [Indexed: 10/28/2022]
Abstract
During morphogenesis tissues significantly remodel by coordinated cell migrations and cell rearrangements. Central to this problem are cell shape changes that are driven by distinct cytoskeletal reorganization responsible for force generation. Calcium is a versatile and universal messenger that is implicated in the regulation of embryonic development. Although calcium transients accrue clearly and more intensely in tissues undergoing rearrangement/migration, it is far from clear what the role of these calcium signals is. Here we summarize the evidence implicating calcium participation in tissue movements, cell shape changes and the reorganization of contractile cytoskeletal elements in developing embryos. We also discuss a novel hypothesis that short-lived calcium spikes are required in cells and tissues undergoing migration and rearrangements as a fine tuning response mechanism to prevent local, abnormally high fluctuations in cytoskeletal activities.
Collapse
Affiliation(s)
- Olga Markova
- IBDML, UMR7288 CNRS-Aix-Marseille Université, Campus de Luminy, Marseille, France.
| | | |
Collapse
|
76
|
Numata T, Sato K, Christmann J, Marx R, Mori Y, Okada Y, Wehner F. The ΔC splice-variant of TRPM2 is the hypertonicity-induced cation channel in HeLa cells, and the ecto-enzyme CD38 mediates its activation. J Physiol 2012; 590:1121-38. [PMID: 22219339 PMCID: PMC3381820 DOI: 10.1113/jphysiol.2011.220947] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 12/31/2011] [Indexed: 12/13/2022] Open
Abstract
Hypertonicity-induced cation channels (HICCs) are key-players in proliferation and apoptosis but their molecular correlate remains obscure. Furthermore, the activation profile of HICCs is not well defined yet. We report here that, in HeLa cells, intracellular adenosine diphosphate ribose (ADPr) and cyclic ADPr (cADPr), as supposed activators of TRPM2, elicited cation currents that were virtually identical to the osmotic activation of HICCs. Silencing of the expression of TRPM2 and of the ecto-enzyme CD38 (as a likely source of ADPr and cADPr) inhibited HICC as well as nucleotide-induced currents and, in parallel, the hypertonic volume response of cells (the regulatory volume increase, RVI) was attenuated. Quantification of intracellular cADPr levels and the systematic application of extra- vs. intracellular nucleotides indicate that the outwardly directed gradient rather than the cellular activity of ADPr and cADPr triggers TRPM2 activation, probably along with a simultaneous biotransformation of nucleotides.Cloning of TRPM2 identified the ΔC-splice variant as the molecular correlate of the HICC, which could be strongly supported by a direct comparison of the respective Ca²⁺ selectivity. Finally, immunoprecipitation and high-resolution FRET/FLIM imaging revealed the interaction of TRPM2 and CD38 in the native as well as in a heterologous (HEK293T) expression system. We propose transport-related nucleotide export via CD38 as a novel mechanism of TRPM2/HICC activation. With the biotransformation of nucleotides running in parallel, continuous zero trans-conditions are achieved which will render the system infinitely sensitive.
Collapse
Affiliation(s)
- Tomohiro Numata
- Department of Cell Physiology, National Institute of Physiological Sciences, Okazaki 444–8585, Japan
| | | | | | | | | | | | | |
Collapse
|
77
|
Schwab A, Nechyporuk-Zloy V, Gassner B, Schulz C, Kessler W, Mally S, Römer M, Stock C. Dynamic redistribution of calcium sensitive potassium channels (hK(Ca)3.1) in migrating cells. J Cell Physiol 2012; 227:686-96. [PMID: 21465474 DOI: 10.1002/jcp.22776] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Calcium-sensitive potassium channels (K(Ca)3.1) are expressed in virtually all migrating cells. Their activity is required for optimal cell migration so that their blockade leads to slowing down. K(Ca)3.1 channels must be inserted into the plasma membrane in order to elicit their physiological function. However, the plasma membrane of migrating cells is subject to rapid recycling by means of endo- and exocytosis. Here, we focussed on the endocytic internalization and the intracellular transport of the human isoform hK(Ca)3.1. A hK(Ca)3.1 channel construct with an HA-tag in the extracellularly located S3-S4 linker was transfected into migrating transformed renal epithelial MDCK-F cells. Channel internalization was visualized and quantified with immunofluorescence and a cell-based ELISA. Movement of hK(Ca)3.1 channel containing vesicles as well as migration of MDCK-F cells were monitored by means of time lapse video microscopy. hK(Ca)3.1 channels are endocytosed during migration. Most of the hK(Ca)3.1 channel containing vesicles are moving at a speed of up to 2 µm/sec in a microtubule-dependent manner towards the front of MDCK-F cells. Our experiments indicate that endocytosis of hK(Ca)3.1 channels is clathrin-dependent since they colocalize with clathrin adaptor proteins and since it is impaired when a C-terminal dileucine motif is mutated. The C-terminal dileucine motif is also important for the subcellular localization of hK(Ca)3.1 channels in migrating cells. Mutated channels are no longer concentrated at the leading edge. We therefore propose that recycling of hK(Ca)3.1 channels contributes to their characteristic subcellular distribution in migrating cells.
Collapse
Affiliation(s)
- Albrecht Schwab
- Institute of Physiology II, Westfälische Wilhelms-Univsität Münster, Münster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Akita T, Fedorovich SV, Okada Y. Ca2+ nanodomain-mediated component of swelling-induced volume-sensitive outwardly rectifying anion current triggered by autocrine action of ATP in mouse astrocytes. Cell Physiol Biochem 2011; 28:1181-90. [PMID: 22179006 DOI: 10.1159/000335867] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2011] [Indexed: 12/20/2022] Open
Abstract
The volume-sensitive outwardly rectifying (VSOR) anion channel provides a major pathway for anion transport during cell volume regulation. It is typically activated in response to cell swelling, but how the channel senses the swelling remains unclear. Meanwhile, we recently found that in mouse astrocytes the channel is activated by an inflammatory chemical mediator, bradykinin, without cell swelling and that the activation is regulated via high concentration regions of intracellular Ca(2+) ([Ca(2+)](i)) in the immediate vicinity of open Ca(2+)-permeable channels, so-called Ca(2+) nanodomains. Here we investigated whether a similar mechanism is involved in the swelling-induced VSOR channel activation in the astrocytes. A hypotonic stimulus (25% reduction in osmolality) caused the [Ca(2+)](i) rises in the astrocytes, and the rises were abolished in the presence of an ATP-degrading enzyme, apyrase (10 U/ml). Application of ATP (100 μM) under isotonic conditions generated the current through VSOR channels via Ca(2+) nanodomains, as bradykinin does. The current induced by the hypotonic stimulus was suppressed by ~40% in the Ca(2+)-depleted condition where the ATP-induced VSOR current was totally prevented. Thus the swelling-induced VSOR channel activation in mouse astrocytes is partly regulated via Ca(2+) nanodomains, whose generation is triggered by an autocrine action of ATP.
Collapse
Affiliation(s)
- Tenpei Akita
- Department of Cell Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan.
| | | | | |
Collapse
|
79
|
Hoffmann EK. Ion channels involved in cell volume regulation: effects on migration, proliferation, and programmed cell death in non adherent EAT cells and adherent ELA cells. Cell Physiol Biochem 2011; 28:1061-78. [PMID: 22178996 DOI: 10.1159/000335843] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2011] [Indexed: 12/26/2022] Open
Abstract
This mini review outlines studies of cell volume regulation in two closely related mammalian cell lines: nonadherent Ehrlich ascites tumour cells (EATC) and adherent Ehrlich Lettre ascites (ELA) cells. Focus is on the regulatory volume decrease (RVD) that occurs after cell swelling, the volume regulatory ion channels involved, and the mechanisms (cellular signalling pathways) that regulate these channels. Finally, I shall also briefly review current investigations in these two cell lines that focuses on how changes in cell volume can regulate cell functions such as cell migration, proliferation, and programmed cell death.
Collapse
Affiliation(s)
- Else Kay Hoffmann
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
80
|
Shlyonsky V, Ben Soussia I, Naeije R, Mies F. Opposing Effects of Bone Morphogenetic Protein-2 and Endothelin-1 on Lung Fibroblast Chloride Currents. Am J Respir Cell Mol Biol 2011; 45:1154-60. [DOI: 10.1165/rcmb.2010-0443oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
81
|
Maillé E, Trinh NTN, Privé A, Bilodeau C, Bissonnette É, Grandvaux N, Brochiero E. Regulation of normal and cystic fibrosis airway epithelial repair processes by TNF-α after injury. Am J Physiol Lung Cell Mol Physiol 2011; 301:L945-55. [DOI: 10.1152/ajplung.00149.2011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic infection and inflammation have been associated with progressive airway epithelial damage in patients with cystic fibrosis (CF). However, the effect of inflammatory products on the repair capacity of respiratory epithelia is unclear. Our objective was to study the regulation of repair mechanisms by tumor necrosis factor-α (TNF-α), a major component of inflammation in CF, in a model of mechanical wounding, in two bronchial cell lines, non-CF NuLi and CF CuFi. We observed that TNF-α enhanced the NuLi and CuFi repair rates. Chronic exposure (24–48 h) to TNF-α augmented this stimulation as well as the migration rate during repair. The cellular mechanisms involved in this stimulation were then evaluated. First, we discerned that TNF-α induced metalloproteinase-9 release, epidermal growth factor (EGF) shedding, and subsequent EGF receptor transactivation. Second, TNF-α-induced stimulation of the NuLi and CuFi wound-closure rates was prevented by GM6001 (metalloproteinase inhibitor), EGF antibody (to titrate secreted EGF), and EGF receptor tyrosine kinase inhibitors. Furthermore, we recently reported a relationship between the EGF response and K+channel function, both controlling bronchial repair. We now show that TNF-α enhances KvLQT1 and KATPcurrents, while their inhibition abolishes TNF-α-induced repair stimulation. These results indicate that the effect of TNF-α is mediated, at least in part, through EGF receptor transactivation and K+channel stimulation. In contrast, cell proliferation during repair was slowed by TNF-α, suggesting that TNF-α could exert contrasting actions on repair mechanisms of CF airway epithelia. Finally, the stimulatory effect of TNF-α on airway wound repair was confirmed on primary airway epithelial cells, from non-CF and CF patients.
Collapse
Affiliation(s)
- Emilie Maillé
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
- Département de médecine and
| | - Nguyen Thu Ngan Trinh
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
- Département de médecine and
| | - Anik Privé
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
| | - Claudia Bilodeau
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
| | - Élyse Bissonnette
- Institut Universitaire de cardiologie et de pneumologie de Québec, Département de médecine, Université Laval, Québec, Québec, Canada
| | - Nathalie Grandvaux
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
- Département de Biochimie, Université de Montréal, Montréal; and
| | - Emmanuelle Brochiero
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM)—Hôtel-Dieu, Montréal
- Département de médecine and
| |
Collapse
|
82
|
Banderali U, Jayanthan A, Hoeksema KA, Narendran A, Giles WR. Ion channels in pediatric CNS Atypical Teratoid/Rhabdoid Tumor (AT/RT) cells: potential targets for novel therapeutic agents. J Neurooncol 2011; 107:111-9. [PMID: 21971736 DOI: 10.1007/s11060-011-0735-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 09/24/2011] [Indexed: 10/17/2022]
Abstract
The central nervous system Atypical Teratoid/Rhabdoid Tumor (CNS AT/RT) is a highly malignant neoplasm that commonly affects infants and young children, and has an extremely poor prognosis. Recently, a small subset of ion channels have been found to be over-expressed in a variety of malignant cells, thus emerging as potential therapeutic targets for difficult to treat tumors. We have studied the electrophysiological properties of AT/RT cell lines with particular attention to cell volume sensitive ion channels (VSC). This class of membrane proteins can play a fundamental role in cellular processes relevant to tumor development. We have found that chloride selective VSCs are particularly active in AT/RT cell lines, compared to non-tumor cells. We evaluated specific inhibitors for activity against chloride selective VSCs and consequently for their ability to inhibit the growth and survival of AT/RT cells in vitro. The results demonstrated that the extent of volume sensitive membrane current inhibition by these agents was correlated with their potency in AT/RT cell growth inhibition in vitro. In addition, we showed that ion channel inhibition enhanced the activity of certain anti-neoplastic agents, suggesting its value in effective drug combination protocols. Results presented provide preliminary in vitro data for possible evaluation of distinct ion channels as plausible therapeutic targets in the treatment of AT/RT.
Collapse
Affiliation(s)
- Umberto Banderali
- Faculty of Kinesiology, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.
| | | | | | | | | |
Collapse
|
83
|
Arcangeli A. Ion channels and transporters in cancer. 3. Ion channels in the tumor cell-microenvironment cross talk. Am J Physiol Cell Physiol 2011; 301:C762-71. [DOI: 10.1152/ajpcell.00113.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The traditional view of cancer as a collection of proliferating cells must be reconsidered, and cancer must be viewed as a “tissue” constituted by both transformed cells and a heterogeneous microenvironment, that tumor cells construct and remodel during multistep tumorigenesis. The “tumor microenvironment” (TM) is formed by mesenchymal, endothelial, and immune cells immersed in a network of extracellular matrix (ECM) proteins and soluble factors. The TM strongly contributes to tumor progression, through long distance, cell-to-cell or cell-to-matrix signals, which influence different aspects of tumor cell behavior. Understanding the relationships among the different components of the cancer tissue is crucial to design and develop new therapeutic strategies. Ion channels are emerging as relevant players in the cross talk between tumor cells and their TM. Ion channels are expressed on tumor cells, as well as in the different cellular components of the TM. In all these cells, ion channels are in a strategic position to sense and transmit extracellular signals into the intracellular machinery. Often, this transmission is mediated by integrin adhesion receptors, which can be functional partners of ion channels since they form molecular complexes with the channel protein in the context of the plasma membrane. The same relevant role is exerted by ion transporters, which also contribute to determine two facets of the cancer tissue: hypoxia and the acidic extracellular pH. On the whole, it is conceivable to prospect the targeting of ion channels for new therapeutic strategies aimed at better controlling the malignant progression of the cancer tissue.
Collapse
Affiliation(s)
- Annarosa Arcangeli
- Department of Experimental Pathology and Oncology, University of Firenze, and Istituto Toscano Tumori, Firenze, Italy
| |
Collapse
|
84
|
Karlsson T, Glogauer M, Ellen RP, Loitto VM, Magnusson KE, Magalhães MAO. Aquaporin 9 phosphorylation mediates membrane localization and neutrophil polarization. J Leukoc Biol 2011; 90:963-73. [PMID: 21873454 DOI: 10.1189/jlb.0910540] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Neutrophils are of prime importance in the host innate defense against invading microorganisms by using two primary mechanisms-locomotion toward and phagocytosis of the prey. Recent research points to pivotal roles for water channels known as AQPs in cell motility. Here, we focused on the role of AQP9 in chemoattractant-induced polarization and migration of primary mouse neutrophils and neutrophil-like HL60 cells. We found that AQP9 is phosphorylated downstream of fMLFR or PMA stimulation in primary human neutrophils. The dynamics of AQP9 were assessed using GFP-tagged AQP9 constructs and other fluorescent markers through various live-cell imaging techniques. Expression of WT or the phosphomimic S11D AQP9 changed cell volume regulation as a response to hyperosmotic changes and enhanced neutrophil polarization and chemotaxis. WT AQP9 and S11D AQP9 displayed a very dynamic distribution at the cell membrane, whereas the phosphorylation-deficient S11A AQP9 failed to localize to the plasma membrane. Furthermore, we found that Rac1 regulated the translocation of AQP9 to the plasma membrane. Our results show that AQP9 plays an active role in neutrophil volume regulation and migration. The display of AQP9 at the plasma membrane depends on AQP9 phosphorylation, which appeared to be regulated through a Rac1-dependent pathway.
Collapse
Affiliation(s)
- Thommie Karlsson
- Division of Medical Microbiology, Department of Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden
| | | | | | | | | | | |
Collapse
|
85
|
Keren K. Cell motility: the integrating role of the plasma membrane. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2011; 40:1013-27. [PMID: 21833780 PMCID: PMC3158336 DOI: 10.1007/s00249-011-0741-0] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 07/24/2011] [Indexed: 11/30/2022]
Abstract
The plasma membrane is of central importance in the motility process. It defines the boundary separating the intracellular and extracellular environments, and mediates the interactions between a motile cell and its environment. Furthermore, the membrane serves as a dynamic platform for localization of various components which actively participate in all aspects of the motility process, including force generation, adhesion, signaling, and regulation. Membrane transport between internal membranes and the plasma membrane, and in particular polarized membrane transport, facilitates continuous reorganization of the plasma membrane and is thought to be involved in maintaining polarity and recycling of essential components in some motile cell types. Beyond its biochemical composition, the mechanical characteristics of the plasma membrane and, in particular, membrane tension are of central importance in cell motility; membrane tension affects the rates of all the processes which involve membrane deformation including edge extension, endocytosis, and exocytosis. Most importantly, the mechanical characteristics of the membrane and its biochemical composition are tightly intertwined; membrane tension and local curvature are largely determined by the biochemical composition of the membrane and the biochemical reactions taking place; at the same time, curvature and tension affect the localization of components and reaction rates. This review focuses on this dynamic interplay and the feedbacks between the biochemical and biophysical characteristics of the membrane and their effects on cell movement. New insight on these will be crucial for understanding the motility process.
Collapse
Affiliation(s)
- Kinneret Keren
- Department of Physics, The Network Biology Research Laboratories and The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, 32000 Haifa, Israel.
| |
Collapse
|
86
|
Magrini E, Szabò I, Doni A, Cibella J, Viola A. Serotonin-mediated tuning of human helper T cell responsiveness to the chemokine CXCL12. PLoS One 2011; 6:e22482. [PMID: 21853036 PMCID: PMC3154189 DOI: 10.1371/journal.pone.0022482] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 06/22/2011] [Indexed: 11/19/2022] Open
Abstract
In addition to its role as neurotransmitter, serotonin (5-HT) is an important modulator of inflammation and immunity. Here, we report novel findings suggesting a 5-HT involvement in T cell migration. In particular, we show that 5-HT tunes the responsiveness of human T lymphocytes to the broadly expressed chemokine CXCL12 in transwell migration assays. By real-time PCR, western blot analysis and electrophysiological patch clamp experiments, we demonstrate that the type 3 5-HT receptor (5-HT3) is functionally expressed in human primary T cells. In addition, specific 5-HT3 receptor agonists selectively decrease T cell migration towards gradients of CXCL12 but not of inflammatory chemokines, such as CCL2 and CCL5. In transmigration experiments, 5-HT3 receptor stimulation reverts the inhibitory effect of endothelial-bound CXCL12 on T cell migration. Our data suggest that the reduced T cell responsiveness to CXCL12 induced by 5-HT may occur to facilitate T cell extravasation and migration into inflamed tissues.
Collapse
Affiliation(s)
- Elena Magrini
- Humanitas Clinical Institute IRCCS, Rozzano, Milan, Italy.
| | | | | | | | | |
Collapse
|
87
|
Abstract
Cell volume homeostasis and its fine-tuning to the specific physiological context at any given moment are processes fundamental to normal cell function. The understanding of cell volume regulation owes much to August Krogh, yet has advanced greatly over the last decades. In this review, we outline the historical context of studies of cell volume regulation, focusing on the lineage started by Krogh, Bodil Schmidt-Nielsen, Hans-Henrik Ussing, and their students. The early work was focused on understanding the functional behaviour, kinetics and thermodynamics of the volume-regulatory ion transport mechanisms. Later work addressed the mechanisms through which cellular signalling pathways regulate the volume regulatory effectors or flux pathways. These studies were facilitated by the molecular identification of most of the relevant channels and transporters, and more recently also by the increased understanding of their structures. Finally, much current research in the field focuses on the most up- and downstream components of these paths: how cells sense changes in cell volume, and how cell volume changes in turn regulate cell function under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- E K Hoffmann
- Section of Cell and Developmental Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
88
|
Bomben VC, Turner KL, Barclay TTC, Sontheimer H. Transient receptor potential canonical channels are essential for chemotactic migration of human malignant gliomas. J Cell Physiol 2011; 226:1879-88. [PMID: 21506118 DOI: 10.1002/jcp.22518] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The majority of malignant primary brain tumors are gliomas, derived from glial cells. Grade IV gliomas, Glioblastoma multiforme, are extremely invasive and the clinical prognosis for patients is dismal. Gliomas utilize a number of proteins and pathways to infiltrate the brain parenchyma including ion channels and calcium signaling pathways. In this study, we investigated the localization and functional relevance of transient receptor potential canonical (TRPC) channels in glioma migration. We show that gliomas are attracted in a chemotactic manner to epidermal growth factor (EGF). Stimulation with EGF results in TRPC1 channel localization to the leading edge of migrating D54MG glioma cells. Additionally, TRPC1 channels co-localize with the lipid raft proteins, caveolin-1 and β-cholera toxin, and biochemical assays show TRPC1 in the caveolar raft fraction of the membrane. Chemotaxis toward EGF was lost when TRPC channels were pharmacologically inhibited or by shRNA knockdown of TRPC1 channels, yet without affecting unstimulated cell motility. Moreover, lipid raft integrity was required for gliomas chemotaxis. Disruption of lipid rafts not only impaired chemotaxis but also impaired TRPC currents in whole cell recordings and decreased store-operated calcium entry as revealed by ratiomeric calcium imaging. These data indicated that TRPC1 channel association with lipid rafts is essential for glioma chemotaxis in response to stimuli, such as EGF.
Collapse
Affiliation(s)
- Valerie C Bomben
- Department of Neurobiology, Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | |
Collapse
|
89
|
Catacuzzeno L, Aiello F, Fioretti B, Sforna L, Castigli E, Ruggieri P, Tata AM, Calogero A, Franciolini F. Serum-activated K and Cl currents underlay U87-MG glioblastoma cell migration. J Cell Physiol 2011; 226:1926-33. [PMID: 21506123 DOI: 10.1002/jcp.22523] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glioblastoma cells in vivo are exposed to a variety of promigratory signals, including undefined serum components that infiltrate into high grade gliomas as result of blood-brain barrier breakdown. Glioblastoma cell migration has been further shown to depend heavily on ion channels activity. We have then investigated the modulatory effects of fetal calf serum (FCS) on ion channels, and their involvement in U87-MG cells migration. Using the perforated patch-clamp technique we have found that, in a subpopulation of cells (42%), FCS induced: (1) an oscillatory activity of TRAM-34 sensitive, intermediate-conductance calcium-activated K (IK(Ca) ) channels, mediated by calcium oscillations previously shown to be induced by FCS in this cell line; (2) a stable activation of a DIDS- and NPPB-sensitive Cl current displaying an outward rectifying instantaneous current-voltage relationship and a slow, voltage-dependent inactivation. By contrast, in another subpopulation of cells (32%) FCS induced a single, transient IK(Ca) current activation, always accompanied by a stable activation of the Cl current. The remaining cells did not respond to FCS. In order to understand whether the FCS-induced ion channel activities are instrumental to promoting cell migration, we tested the effects of TRAM-34 and DIDS on the FCS-induced U87-MG cell migration using transwell migration assays. We found that these inhibitors were able to markedly reduce U87-MG cell migration in the presence of FCS, and that their co-application resulted in an almost complete arrest of migration. It is concluded that the modulation of K and Cl ion fluxes is essential for the FCS-induced glioblastoma cell migration.
Collapse
Affiliation(s)
- Luigi Catacuzzeno
- Dipartimento di Biologia Cellulare e Ambientale, Universita' di Perugia, Perugia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Cuddapah VA, Sontheimer H. Ion channels and transporters [corrected] in cancer. 2. Ion channels and the control of cancer cell migration. Am J Physiol Cell Physiol 2011; 301:C541-9. [PMID: 21543740 DOI: 10.1152/ajpcell.00102.2011] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A hallmark of high-grade cancers is the ability of malignant cells to invade unaffected tissue and spread disease. This is particularly apparent in gliomas, the most common and lethal type of primary brain cancer affecting adults. Migrating cells encounter restricted spaces and appear able to adjust their shape to accommodate to narrow extracellular spaces. A growing body of work suggests that cell migration/invasion is facilitated by ion channels and transporters. The emerging concept is that K(+) and Cl(-) function as osmotically active ions, which cross the plasma membrane in concert with obligated water thereby adjusting a cell's shape and volume. In glioma cells Na(+)-K(+)-Cl(-) cotransporters (NKCC1) actively accumulate K(+) and Cl(-), establishing a gradient for KCl efflux. Ca(2+)-activated K(+) channels and voltage-gated Cl(-) channels are largely responsible for effluxing KCl promoting hydrodynamic volume changes. In other cancers, different K(+) or even Na(+) channels may function in concert with a variety of Cl(-) channels to support similar volume changes. Channels involved in migration are frequently regulated by Ca(2+) signaling, most likely coupling extracellular stimuli to cell migration. Importantly, the inhibition of ion channels and transporters appears to be clinically relevant for the treatment of cancer. Recent preclinical data indicates that inhibition of NKCC1 with an FDA-approved drug decreases neoplastic migration. Additionally, ongoing clinical trials demonstrate that an inhibitor of chloride channels may be a therapy for the treatment of gliomas. Data reviewed here strongly indicate that ion channels are a promising target for the development of novel therapeutics to combat cancer.
Collapse
Affiliation(s)
- Vishnu Anand Cuddapah
- Department of Neurobiology and Center for Glial Biology in Medicine, University of Alabama at Birmingham, USA
| | | |
Collapse
|
91
|
Shao Z, Makinde TO, Agrawal DK. Calcium-activated potassium channel KCa3.1 in lung dendritic cell migration. Am J Respir Cell Mol Biol 2011; 45:962-8. [PMID: 21493782 DOI: 10.1165/rcmb.2010-0514oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Migration to draining lymph nodes is a critical requirement for dendritic cells (DCs) to control T-cell-mediated immunity. The calcium-activated potassium channel KCa3.1 has been shown to be involved in regulating cell migration in multiple cell types. In this study, KCa3.1 expression and its functional role in lung DC migration were examined. Fluorescence-labeled antigen was intranasally delivered into mouse lungs to label lung Ag-carrying DCs. Lung CD11c(high)CD11b(low) and CD11c(low)CD11b(high) DCs from PBS-treated and ovalbumin (OVA)-sensitized mice were sorted using MACS and FACS. Indo-1 and DiBAC4(3) were used to measure intracellular Ca(2+) and membrane potential, respectively. The mRNA expression of KCa3.1 was examined using real-time PCR. Expression of KCa3.1 protein and CCR7 was measured using flow cytometry. Migration of two lung DC subsets to lymphatic chemokines was examined using TransWell in the absence or presence of the KCa3.1 blocker TRAM-34. OVA sensitization up-regulated mRNA and protein expression of KCa3.1 in lung DCs, with a greater response by the CD11c(high)CD11b(low) than CD11c(low)CD11b(high) DCs. Although KCa3.1 expression in Ag-carrying DCs was higher than that in non-Ag-carrying DCs in OVA-sensitized mice, the difference was not as prominent. However, Ag-carrying lung DCs expressed significantly higher CCR7 than non-Ag-carrying DCs. CCL19, CCL21, and KCa3.1 activator 1-EBIO induced an increase in intracellular calcium in both DC subsets. In addition, 1-EBIO-induced calcium increase was suppressed by TRAM-34. In vitro blockade of KCa3.1 with TRAM-34 impaired CCL19/CCL21-induced transmigration. In conclusion, KCa3.1 expression in lung DCs is up-regulated by OVA sensitization in both lung DC subsets, and KCa3.1 is involved in lung DC migration to lymphatic chemokines.
Collapse
Affiliation(s)
- Zhifei Shao
- Center for Clinical & Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA
| | | | | |
Collapse
|
92
|
Waschk DEJ, Fabian A, Budde T, Schwab A. Dual-color quantum dot detection of a heterotetrameric potassium channel (hKCa3.1). Am J Physiol Cell Physiol 2011; 300:C843-9. [PMID: 21228320 DOI: 10.1152/ajpcell.00053.2010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Potassium channels play a key role in establishing the cell membrane potential and are expressed ubiquitously. Today, more than 70 mammalian K(+) channel genes are known. The diversity of K(+) channels is further increased by the fact that different K(+) channel family members may assemble to form heterotetramers. We present a method based on fluorescence microscopy to determine the subunit composition of a tetrameric K(+) channel. We generated artificial "heteromers" of the K(+) channel hK(Ca)3.1 by coexpressing two differently tagged hK(Ca)3.1 constructs containing either an extracellular hemagglutinin (HA) or an intracellular V5 epitope. hK(Ca)3.1 channel subunits were detected in the plasma membrane of MDCK-F cells or HEK293 cells by labeling the extra- and intracellular epitopes with differently colored quantum dots (QDs). As previously shown for the extracellular part of hK(Ca)3.1 channels, its intracellular domain can also bind only one QD label at a time. When both channel subunits were coexpressed, 27.5 ± 1.8% and 24.9 ± 2.1% were homotetramers consisting of HA- and V5-tagged subunits, respectively. 47.6 ± 3.2% of the channels were heteromeric and composed of both subunits. The frequency distribution of HA- and V5-tagged homo- and heteromeric hK(Ca)3.1 channels is reminiscent of the binomial distribution (a + b)(2) = a(2) + 2ab + b(2). Along these lines, our findings are consistent with the notion that hK(Ca)3.1 channels are assembled from two homomeric dimers and not randomly from four independent subunits. We anticipate that our technique will be applicable to other heteromeric membrane proteins, too.
Collapse
|
93
|
Potier M, Chantome A, Joulin V, Girault A, Roger S, Besson P, Jourdan ML, LeGuennec JY, Bougnoux P, Vandier C. The SK3/K(Ca)2.3 potassium channel is a new cellular target for edelfosine. Br J Pharmacol 2011; 162:464-79. [PMID: 20955368 PMCID: PMC3031066 DOI: 10.1111/j.1476-5381.2010.01044.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 08/31/2010] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The 1-O-octadecyl-2-O-methyl-sn-glycero-3-phosphocholine (edelfosine) is an ether-linked phospholipid with promising anti-cancer properties but some side effects that preclude its full clinical therapeutic exploitation. We hypothesized that this lipid could interact with plasma membrane ion channels and modulate their function. EXPERIMENTAL APPROACH Using cell migration-proliferation assays, patch clamp, spectrofluorimetry and ¹²⁵I-Apamin binding experiments, we studied the effects of edelfosine on the migration of breast cancer MDA-MB-435s cells, mediated by the small conductance Ca²(+) -activated K(+) channel, SK3/K(Ca)2.3. KEY RESULTS Edelfosine (1 µM) caused plasma membrane depolarization by substantially inhibiting activity of SK3/K(Ca)2.3 channels, which we had previously demonstrated to play an important role in cancer cell migration. Edelfosine did not inhibit ¹²⁵I-Apamin binding to this SK(Ca) channel; rather, it reduced the calcium sensitivity of SK3/K(Ca)2.3 channel and dramatically decreased intracellular Ca²(+) concentration, probably by insertion in the plasma membrane, as suggested by proteinase K experiments. Edelfosine reduced cell migration to the same extent as known SK(Ca) channel blockers. In contrast, K+ channel openers prevented edelfosine-induced anti-migratory effects. SK3 protein knockdown decreased cell migration and totally abolished the effect of edelfosine on MDA-MB-435s cell migration. In contrast, transient expression of SK3/K(Ca)2.3 protein in a SK3/K(Ca)2.3-deficient cell line increased cell migration and made these cells responsive to edelfosine. CONCLUSIONS AND IMPLICATIONS Our data clearly establish edelfosine as an inhibitor of cancer cell migration by acting on SK3/K(Ca)2.3 channels and provide insights into the future development of a new class of migration-targeted, anti-cancer agents.
Collapse
Affiliation(s)
- M Potier
- Inserm, U, Université François Rabelais, Tours, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Seidler U, Song P, Xiao F, Riederer B, Bachmann O, Chen M. Recent advances in the molecular and functional characterization of acid/base and electrolyte transporters in the basolateral membranes of gastric and duodenal epithelial cells. Acta Physiol (Oxf) 2011; 201:3-20. [PMID: 20331540 DOI: 10.1111/j.1748-1716.2010.02107.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
All segments of the gastrointestinal tract are comprised of an elaborately folded epithelium that expresses a variety of cell types and performs multiple secretory and absorptive functions. While the apical membrane expresses the electrolyte transporters that secrete or absorb electrolytes and water, basolateral transporters regulate the secretory or absorptive rates. During gastric acid formation, Cl⁻/HCO₃⁻ and Na(+) /H(+) exchange and other transporters secure Cl⁻ re-supply as well as pH and volume regulation. Gastric surface cells utilize ion transporters to secrete HCO₃⁻, maintain pH(i) during a luminal acid load and repair damaged surface areas during the process of epithelial restitution. Na(+)/H(+) exchange and Na(+)/HCO₃⁻ cotransport serve basolateral acid/base import for gastroduodenal HCO₃⁻ secretion. The gastric and duodenal epithelium also absorbs salt and water. Recent molecular information on novel ion transporters expressed in the gastric and duodenal epithelium has exploded; however, a function has not been found yet for all transporters. The purpose of this review is to summarize current knowledge on the molecular identity and cellular function of basolateral ion transporters in the gastric and duodenal epithelium.
Collapse
Affiliation(s)
- U Seidler
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Germany.
| | | | | | | | | | | |
Collapse
|
95
|
Chemotaxis of MDCK-F cells toward fibroblast growth factor-2 depends on transient receptor potential canonical channel 1. Pflugers Arch 2010; 461:295-306. [PMID: 21120665 DOI: 10.1007/s00424-010-0901-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 11/02/2010] [Accepted: 11/03/2010] [Indexed: 01/25/2023]
Abstract
Movement toward the source of a chemoattractant gradient is a basic cellular property in health and disease. Enhanced migration during metastasis involves deregulated growth factor signaling. Growth factor stimulation and cell migration converge both on the important second messenger Ca(2+). To date, the molecular identification of Ca(2+) entry pathways activated by growth factors during chemotaxis is still an open issue. We investigated the involvement of the nonselective Ca(2+) channel TRPC1 (transient receptor potential canonical 1) in FGF-2 guided chemotaxis by means of time-lapse video microscopy and by functional Ca(2+) measurements. To specifically address TRPC1 function in transformed MDCK cells we altered the expression levels by siRNA or overexpression. We report that TRPC1 channels are required for the orientation of transformed MDCK cells in FGF-2 gradients because TRPC1 knockdown or pharmacological blockade prevented chemotaxis. Stimulation with FGF-2 triggered an immediate Ca(2+) influx via TRPC1 channels that depended on phospholipase C and phosphatidylinositol 3-kinase signaling. Impeding this Ca(2+) influx abolished chemotaxis toward FGF-2. This functional connection correlated with clustering of FGF receptors and TRPC1 channels as was observed by immunolabeling. These findings show the important interplay between growth factor signaling and Ca(2+) influx in chemotaxis.
Collapse
|
96
|
Ding T, Gu F, Fu L, Ma YJ. Aquaporin-4 in glioma invasion and an analysis of molecular mechanisms. J Clin Neurosci 2010; 17:1359-61. [DOI: 10.1016/j.jocn.2010.02.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 02/04/2010] [Accepted: 02/05/2010] [Indexed: 12/23/2022]
|
97
|
Kirk KL. CFTR channels and wound healing. Focus on “Cystic fibrosis transmembrane conductance regulator is involved in airway epithelial wound repair”. Am J Physiol Cell Physiol 2010; 299:C888-90. [DOI: 10.1152/ajpcell.00313.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Kevin L. Kirk
- Department of Physiology and Biophysics and Department of Neurobiology, Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
98
|
Kraemer BF, Borst O, Gehring EM, Schoenberger T, Urban B, Ninci E, Seizer P, Schmidt C, Bigalke B, Koch M, Martinovic I, Daub K, Merz T, Schwanitz L, Stellos K, Fiesel F, Schaller M, Lang F, Gawaz M, Lindemann S. PI3 kinase-dependent stimulation of platelet migration by stromal cell-derived factor 1 (SDF-1). J Mol Med (Berl) 2010; 88:1277-88. [PMID: 20852838 DOI: 10.1007/s00109-010-0680-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Revised: 08/10/2010] [Accepted: 08/12/2010] [Indexed: 01/02/2023]
Abstract
Platelets have been regarded as static cells that do not move once they adhere to a matrix. The present study explored, whether platelets are able to migrate. In contrast to the current opinion, we found that platelets were mobile, able to migrate over a surface, and transmigrate through a transwell membrane and endothelium toward a source of stromal cell-derived factor 1 (SDF-1). Platelet migration was stimulated by SDF-1, which led to the downstream activation and phosphorylation of Wiskott-Aldrich syndrome protein. SDF-1 signaling and subsequent platelet migration could be inhibited by CXCR4-receptor blocker AMD3100, pertussis toxin, inhibition of phosphoinositol 3-kinase (PI3 kinase) with LY294002 or wortmannin, and disruption of actin polymerization with cytochalasin B. The potential of platelets to migrate in an SDF-1-mediated fashion may redefine the role of platelets in the pathophysiology of vascular inflammation, subsequent atherosclerotic degeneration, and vascular regeneration.
Collapse
Affiliation(s)
- Bjoern F Kraemer
- Abteilung III, Kardiologie und Kreislauferkrankungen, Medizinische Klinik, Universitätsklinikum Tübingen, Otfried-Müller-Strasse 10, 72076, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Klausen TK, Preisler S, Pedersen SF, Hoffmann EK. Monovalent ions control proliferation of Ehrlich Lettre ascites cells. Am J Physiol Cell Physiol 2010; 299:C714-25. [DOI: 10.1152/ajpcell.00445.2009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Channels and transporters of monovalent ions are increasingly suggested as putative anticarcinogenic targets. However, the mechanisms involved in modulation of proliferation by monovalent ions are poorly understood. Here, we investigated the role of K+, Na+, and Cl− ions for the proliferation of Ehrlich Lettre ascites (ELA) cells. We measured the intracellular concentration of each ion in G0, G1, and S phases of the cell cycle following synchronization by serum starvation and release. We show that intracellular concentrations and content of Na+ and Cl− were reduced in the G0–G1 phase transition, followed by an increased content of both ions in S phase concomitant with water uptake. The effect of substituting extracellular monovalent ions was investigated by bromodeoxyuridine incorporation and showed marked reduction after Na+ and Cl− substitution. In spectrofluorometric measurements with the pH-sensitive dye BCECF, substitution of Na+ was observed to upregulate the activity of the Na+/H+ exchanger NHE1 as well as of Na+-independent acid extrusion mechanisms, facilitating intracellular pH (pHi) recovery after acid loading and increasing pHi. Results using the potential sensitive dye DiBaC4( 3 ) showed a reduced Cl− conductance in S compared with G1 followed by transmembrane potential ( Em) hyperpolarization in S. Cl− substitution by impermeable anions strongly inhibited proliferation and increased free, intracellular Ca2+ ([Ca2+]i), whereas a more permeable anion had little effect. Western blots showed reduced chloride intracellular channel CLIC1 and chloride channel ClC-2 expression in the plasma membrane in S compared with G1. Our results suggest that Na+ regulates ELA cell proliferation by regulating intracellular pH while Cl− may regulate proliferation by fine-tuning of Em in S phase and altered Ca2+ signaling.
Collapse
Affiliation(s)
| | - Sarah Preisler
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Else Kay Hoffmann
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
100
|
Schiller KR, Maniak PJ, O'Grady SM. Cystic fibrosis transmembrane conductance regulator is involved in airway epithelial wound repair. Am J Physiol Cell Physiol 2010; 299:C912-21. [PMID: 20686068 DOI: 10.1152/ajpcell.00215.2010] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The role of cystic fibrosis (CF) transmembrane conductance regulator (CFTR) in airway epithelial wound repair was investigated using normal human bronchial epithelial (NHBE) cells and a human airway epithelial cell line (Calu-3) of serous gland origin. Measurements of wound repair were performed using continuous impedance sensing to determine the time course for wound closure. Control experiments showed that the increase in impedance corresponding to cell migration into the wound was blocked by treatment with the actin polymerization inhibitor, cytochalasin D. Time lapse imaging revealed that NHBE and Calu-3 cell wound closure was dependent on cell migration, and that movement occurred as a collective sheet of cells. Selective inhibition of CFTR activity with CFTR(inh)-172 or short hairpin RNA silencing of CFTR expression produced a significant delay in wound repair. The CF cell line UNCCF1T also exhibited significantly slower migration than comparable normal airway epithelial cells. Inhibition of CFTR-dependent anion transport by treatment with CFTR(inh)-172 slowed wound closure to the same extent as silencing CFTR protein expression, indicating that ion transport by CFTR plays a critical role in migration. Moreover, morphologic analysis of migrating cells revealed that CFTR inhibition or silencing significantly reduced lamellipodia protrusion. These findings support the conclusion that CFTR participates in airway epithelial wound repair by a mechanism involving anion transport that is coupled to the regulation of lamellipodia protrusion at the leading edge of the cell.
Collapse
Affiliation(s)
- Katherine R Schiller
- Graduate Program in Comparative Molecular Biosciences, University of Minnesota, St. Paul, Minnesota 55108, USA
| | | | | |
Collapse
|