51
|
Bartsch I, Willbold E, Rosenhahn B, Witte F. Non-invasive pH determination adjacent to degradable biomaterials in vivo. Acta Biomater 2014; 10:34-9. [PMID: 24021229 DOI: 10.1016/j.actbio.2013.08.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/01/2013] [Accepted: 08/29/2013] [Indexed: 01/07/2023]
Abstract
An appropriate pH level is an important prerequisite for the physiologal functioning of cells and tissues. Changes in the extracellular pH often lead to specific cellular reactions and an altered metabolism of cells and tissues influences the extracellular pH range. Thus a method to monitor the extracellular pH is a valuable tool to track specific tissue reactions. In this article we describe a method for the determination of the pH range adjacent to degradable biomaterials using wireless in vivo imaging. Using hairless but immunocompetent mice the fluorophor 5-(6)-carboxy SNARF-1 and the in vivo fluorescence and multispectral acquisition and analysis system Maestro it is possible to track shifts in pH in small living animals over a longer period of time. This method is especially suitable for studies which focus on the interaction of degrading biomaterials with their adjacent tissues.
Collapse
Affiliation(s)
- Ivonne Bartsch
- Laboratory for Biomechanics and Biomaterials, Department of Orthopaedic Surgery, Hannover Medical School, Anna-von-Borries-Straße 1-7, 30625 Hannover, Germany; CrossBIT, Center for Biocompatibility and Implant-Immunology, Department of Orthopaedic Surgery, Hannover Medical School, Feodor-Lynen-Straße 31, 30625 Hannover, Germany
| | | | | | | |
Collapse
|
52
|
Anoctamin 1 induces calcium-activated chloride secretion and proliferation of renal cyst-forming epithelial cells. Kidney Int 2013; 85:1058-67. [PMID: 24152967 DOI: 10.1038/ki.2013.418] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 08/20/2013] [Accepted: 08/22/2013] [Indexed: 01/08/2023]
Abstract
Polycystic kidney diseases are characterized by multiple bilateral renal cysts that gradually enlarge and lead to a decline in renal function. Cyst enlargement is driven by transepithelial chloride secretion, stimulated by enhanced levels of cyclic adenosine monophosphate, which activates apical cystic fibrosis transmembrane conductance regulator chloride channels. However, chloride secretion by calcium-dependent chloride channels, activated through stimulation of purinergic receptors, also has a major impact. To identify the molecular basis of calcium-dependent chloride secretion in cyst expansion, we determined the role of anoctamin 1 and 6, two recently discovered calcium-activated chloride channels both of which are expressed in epithelial cells. We found that anoctamin 1, which plays a role in epithelial fluid secretion and proliferation, is strongly expressed in principal-like MDCK cells (PLCs) forming cysts within a collagen matrix, in an embryonic kidney cyst model, and in human autosomal dominant polycystic kidney disease tissue. Knockdown of anoctamin 1 but not anoctamin 6 strongly diminished the calcium-dependent chloride secretion of PLCs. Moreover, two inhibitors of anoctamin ion channels, tannic acid and a more selective inhibitor of anoctamin 1, significantly inhibited PLC cyst growth and cyst enlargement in an embryonic kidney cyst model. Knockdown of ANO1 by morpholino analogs also attenuated embryonic cyst growth. Thus, calcium-activated chloride secretion by anoctamin 1 appears to be a crucial component of renal cyst growth.
Collapse
|
53
|
Pedersen SF, Hoffmann EK, Novak I. Cell volume regulation in epithelial physiology and cancer. Front Physiol 2013; 4:233. [PMID: 24009588 PMCID: PMC3757443 DOI: 10.3389/fphys.2013.00233] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Accepted: 08/09/2013] [Indexed: 12/21/2022] Open
Abstract
The physiological function of epithelia is transport of ions, nutrients, and fluid either in secretory or absorptive direction. All of these processes are closely related to cell volume changes, which are thus an integrated part of epithelial function. Transepithelial transport and cell volume regulation both rely on the spatially and temporally coordinated function of ion channels and transporters. In healthy epithelia, specific ion channels/transporters localize to the luminal and basolateral membranes, contributing to functional epithelial polarity. In pathophysiological processes such as cancer, transepithelial and cell volume regulatory ion transport are dys-regulated. Furthermore, epithelial architecture and coordinated ion transport function are lost, cell survival/death balance is altered, and new interactions with the stroma arise, all contributing to drug resistance. Since altered expression of ion transporters and channels is now recognized as one of the hallmarks of cancer, it is timely to consider this especially for epithelia. Epithelial cells are highly proliferative and epithelial cancers, carcinomas, account for about 90% of all cancers. In this review we will focus on ion transporters and channels with key physiological functions in epithelia and known roles in the development of cancer in these tissues. Their roles in cell survival, cell cycle progression, and development of drug resistance in epithelial cancers will be discussed.
Collapse
Affiliation(s)
- Stine F Pedersen
- Department of Biology, University of Copenhagen Copenhagen, Denmark
| | | | | |
Collapse
|
54
|
Intracellular and extracellular pH and Ca are bound to control mitosis in the early sea urchin embryo via ERK and MPF activities. PLoS One 2013; 8:e66113. [PMID: 23785474 PMCID: PMC3681939 DOI: 10.1371/journal.pone.0066113] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 05/01/2013] [Indexed: 11/19/2022] Open
Abstract
Studies aiming to predict the impact on marine life of ocean acidification and of altered salinity have shown altered development in various species including sea urchins. We have analyzed how external Na, Ca, pH and bicarbonate control the first mitotic divisions of sea urchin embryos. Intracellular free Ca (Cai) and pH (pHi) and the activities of the MAP kinase ERK and of MPF regulate mitosis in various types of cells including oocytes and early embryos. We found that intracellular acidification of fertilized eggs by Na-acetate induces a huge activation of ERK at time of mitosis. This also stops the cell cycle and leads to cell death, which can be bypassed by treatment with the MEK inhibitor U0126. Similar intracellular acidification induced in external medium containing low sodium or 5-(N-Methyl-N-isobutyl) amiloride, an inhibitor of the Na+/H+ exchanger, also stops the cell cycle and leads to cell death. In that case, an increase in Cai and in the phosphorylation of tyr-cdc2 occurs during mitosis, modifications that depend on external Ca. Our results indicate that the levels of pHi and Cai determine accurate levels of Ptyr-Cdc2 and P-ERK capable of ensuring progression through the first mitotic cycles. These intracellular parameters rely on external Ca, Na and bicarbonate, alterations of which during climate changes could act synergistically to perturb the early marine life.
Collapse
|
55
|
Dry H, Jorgenson K, Ando W, Hart DA, Frank CB, Sen A. Effect of calcium on the proliferation kinetics of synovium-derived mesenchymal stromal cells. Cytotherapy 2013; 15:805-19. [PMID: 23477785 DOI: 10.1016/j.jcyt.2013.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 12/31/2012] [Accepted: 01/18/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Synovium-derived mesenchymal stromal cells (S-MSCs) have potential utility in clinical joint repair applications. However, their scarcity in tissues means S-MSCs cannot be isolated in large quantities and need to be expanded in culture. Because synovial tissues in vivo are exposed to higher calcium (Ca(2+)) levels than typically found in culture media, this study examined the impact of Ca(2+) supplementation on the rate of S-MSC proliferation in culture. METHODS S-MSCs were serially cultured with or without Ca(2+) supplementation. The effect of inhibiting Ca(2+) uptake was assessed using Ca(2+) channel blockers. After extended exposure to elevated Ca(2+) concentrations, S-MSCs were characterized by evaluating surface marker profiles, performing reverse transcriptase quantitative polymerase chain reaction and carrying out tri-lineage differentiation assays. RESULTS Elevated Ca(2+) concentrations resulted in enhanced S-MSC proliferation. Peak growth occurred at 5.0 mmol/L Ca(2+), with an average fold increase of 4.52 ± 0.65 per passage over 8 passages compared with 2.03 ± 0.46 in un-supplemented medium. Proliferation was inhibited by Ca(2+) channel blockers. Ca(2+)-supplemented cells showed enhanced capacity toward osteogenesis (17.82 ± 4.21 μg Ca(2+) deposited/sample vs. 12.70 ± 2.11 μg Ca(2+) deposited/sample) and adipogenesis (0.47 ± 0.04 mg oil red O/sample vs. 0.352 ± 0.005 mg oil red O/sample) and retained their capacity to undergo chondrogenesis (1.37 ± 0.07 μg glycosaminoglycan/pellet vs. 1.33 ± 0.17 μg glycosaminoglycan/pellet). S-MSCs cultured in elevated Ca(2+) expressed enhanced messenger RNA levels for SOX-9 and peroxisome proliferator activated receptor gamma and depressed levels for collagen I. CONCLUSIONS S-MSC sensitivity to Ca(2+) has not been reported previously. These findings indicate that S-MSC population expansion rates may be up-regulated by Ca(2+) supplementation without compromising defining cell characteristics. This study exemplifies the need to consider medium composition when culturing stem cells.
Collapse
Affiliation(s)
- Helen Dry
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
56
|
Schönichen A, Webb BA, Jacobson MP, Barber DL. Considering protonation as a posttranslational modification regulating protein structure and function. Annu Rev Biophys 2013; 42:289-314. [PMID: 23451893 DOI: 10.1146/annurev-biophys-050511-102349] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Posttranslational modification is an evolutionarily conserved mechanism for regulating protein activity, binding affinity, and stability. Compared with established posttranslational modifications such as phosphorylation or ubiquitination, posttranslational modification by protons within physiological pH ranges is a less recognized mechanism for regulating protein function. By changing the charge of amino acid side chains, posttranslational modification by protons can drive dynamic changes in protein conformation and function. Addition and removal of a proton is rapid and reversible and, in contrast to most other posttranslational modifications, does not require an enzyme. Signaling specificity is achieved by only a minority of sites in proteins titrating within the physiological pH range. Here, we examine the structural mechanisms and functional consequences of proton posttranslational modification of pH-sensing proteins regulating different cellular processes.
Collapse
Affiliation(s)
- André Schönichen
- Department of Cell and Tissue Biology, University of California, San Francisco, USA
| | | | | | | |
Collapse
|
57
|
Park JY, Choi HW, Choi DL, Jang SJ, Kim JH, Lee JH, Choo DJ, Kim J, Lee KT, Lee JY. Evaluation of T-Type Calcium Channel Blockers against Human Pancreatic MIA PaCa-2 Carcinoma Xenografts. B KOREAN CHEM SOC 2013. [DOI: 10.5012/bkcs.2013.34.2.482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
58
|
Neural progenitor cells regulate capillary blood flow in the postnatal subventricular zone. J Neurosci 2013; 32:16435-48. [PMID: 23152626 DOI: 10.1523/jneurosci.1457-12.2012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the postnatal subventricular zone (SVZ), S phase entry of neural progenitor cells (NPCs) correlates with a local increase in blood flow. However, the cellular mechanism controlling this hemodynamic response remains unknown. We show that a subpopulation of SVZ cells, astrocyte-like cells or B-cells, sends projections ensheathing pericytes on SVZ capillaries in young mice. We examined whether calcium increases in pericytes or B-cells led to a vascular response in acute slices using the P2Y(2/4) receptor (P2Y(2/4)R) agonist UTP, electrical stimulation, or transgenic mice expressing exogenous Gq-coupled receptors (MrgA1) in B-cells. UTP increased calcium in pericytes leading to capillary constrictions. Electrical stimulation induced calcium propagation in SVZ cells followed by capillary constrictions involving purinergic receptors. In transgenic mice, selective calcium increases in B-cells induced P2Y(2/4)R-dependent capillary constrictions, suggesting that B-cells release ATP activating purinergic receptors on pericytes. Interestingly, in the presence of a P2Y(2/4)R blocker, dilation was observed. Intraventricular UTP injection transiently decreased blood flow monitored in vivo using laser Doppler flowmetry. Using neonatal electroporation, we expressed MrgA1 in slow cycling radial glia-derived B1 cells, i.e., NPCs. Intraventricular injection of an MrgA1 ligand increased blood flow in the SVZ. Thus, upon intracellular calcium increases B-cells/NPCs release ATP and vasodilating factors that activate purinergic receptors on pericytes triggering a vascular response and blood flow increase in vivo. Considering that NPCs receive signals from other SVZ cells, these findings further suggest that NPCs act as transducers of neurometabolic coupling in the SVZ.
Collapse
|
59
|
Zhang YP, Wang WL, Liu J, Li WB, Bai LL, Yuan YD, Song SX. Plasminogen activator inhibitor-1 promotes the proliferation and inhibits the apoptosis of pulmonary fibroblasts by Ca2+ signaling. Thromb Res 2013; 131:64-71. [DOI: 10.1016/j.thromres.2012.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Revised: 08/18/2012] [Accepted: 09/03/2012] [Indexed: 01/21/2023]
|
60
|
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: 135] [Impact Index Per Article: 10.4] [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
|
61
|
Madsen CP, Klausen TK, Fabian A, Hansen BJ, Pedersen SF, Hoffmann EK. On the role of TRPC1 in control of Ca2+ influx, cell volume, and cell cycle. Am J Physiol Cell Physiol 2012; 303:C625-34. [PMID: 22744003 DOI: 10.1152/ajpcell.00287.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ca(+) signaling plays a crucial role in control of cell cycle progression, but the understanding of the dynamics of Ca(2+) influx and release of Ca(2+) from intracellular stores during the cell cycle is far from complete. The aim of the present study was to investigate the role of the free extracellular Ca(2+) concentration ([Ca(2+)](o)) in cell proliferation, the pattern of changes in the free intracellular Ca(2+) concentration ([Ca(2+)](i)) during cell cycle progression, and the role of the transient receptor potential (TRP)C1 in these changes as well as in cell cycle progression and cell volume regulation. In Ehrlich Lettré Ascites (ELA) cells, [Ca(2+)](i) decreased significantly, and the thapsigargin-releasable Ca(2+) pool in the intracellular stores increased in G(1) as compared with G(0). Store-depletion-operated Ca(2+) entry (SOCE) and TRPC1 protein expression level were both higher in G(1) than in G(0) and S phase, in parallel with a more effective volume regulation after swelling [regulatory volume decrease (RVD)] in G(1) as compared with S phase. Furthermore, reduction of [Ca(2+)](o), as well as two unspecific SOCE inhibitors, 2-APB (2-aminoethyldiphenyl borinate) and SKF96365 (1-(β-[3-(4-methoxy-phenyl)propoxyl-4-methoxyphenethyl)1H-imidazole-hydrochloride), inhibited ELA cell proliferation. Finally, Madin-Darby canine kidney cells in which TRPC1 was stably silenced [TRPC1 knockdown (TRPC1-KD) MDCK] exhibited reduced SOCE, slower RVD, and reduced cell proliferation compared with mock controls. In conclusion, in ELA cells, SOCE and TRPC1 both seem to be upregulated in G(1) as compared with S phase, concomitant with an increased rate of RVD. Furthermore, TRPC1-KD MDCK cells exhibit decreased SOCE, decreased RVD, and decreased proliferation, suggesting that, at least in certain cell types, TRPC1 is regulated during cell cycle progression and is involved in SOCE, RVD, and cell proliferation.
Collapse
Affiliation(s)
- C P Madsen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
62
|
Kang HB, Rim HK, Park JY, Choi HW, Choi DL, Seo JH, Chung KS, Huh G, Kim J, Choo DJ, Lee KT, Lee JY. In vivo evaluation of oral anti-tumoral effect of 3,4-dihydroquinazoline derivative on solid tumor. Bioorg Med Chem Lett 2011; 22:1198-201. [PMID: 22177784 DOI: 10.1016/j.bmcl.2011.11.083] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 11/17/2011] [Accepted: 11/19/2011] [Indexed: 11/25/2022]
Abstract
An extension of our previously reported 3,4-dihydroquinazoline derivative is investigated. Oral anti-tumoral activity of 3,4-dihydroquinazoline derivative (KYS05090) as potent and selective T-type calcium channel blocker was in vivo evaluated against A549 xenograft in BALB/c(nu/nu) nude mice. The rate of tumor volume increment in mouse model with KYS05090-treated group was remarkably slower than that of control group. With respect to tumor weight, it exhibited 60% and 67% tumor growth inhibition through oral administration of 1 and 5mg/kg of bodyweight, respectively, compared to control and was more potent than paclitaxel (53%). In addition, KYS05090 (10 and 50mg/kg, po) was found to have a marked analgesic effect in acetic acid-induced writhing test, whereas it did not show any effect on hot plate test.
Collapse
Affiliation(s)
- Han Byul Kang
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Alvarez-Baron CP, Jonsson P, Thomas C, Dryer SE, Williams C. The two-pore domain potassium channel KCNK5: induction by estrogen receptor alpha and role in proliferation of breast cancer cells. Mol Endocrinol 2011; 25:1326-36. [PMID: 21680658 DOI: 10.1210/me.2011-0045] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The growth of many human breast tumors requires the proliferative effect of estrogen acting via the estrogen receptor α (ERα). ERα signaling is therefore a clinically important target for breast cancer prevention and therapeutics. Although extensively studied, the mechanism by which ERα promotes proliferation remains to be fully established. We observed an up-regulation of transcript encoding the pH-sensitive two-pore domain potassium channel KCNK5 in a screen for genes stimulated by 17β-estradiol (E2) in the ERα(+) breast cancer cell lines MCF-7 and T47D. KCNK5 mRNA increased starting 1 h after the onset of E2 treatment, and protein levels followed after 12 h. Estrogen-responsive elements are found in the enhancer region of KCNK5, and chromatin immunoprecipitation assays revealed binding of ERα to the KCNK5 enhancer in E2-treated MCF-7 cells. Cells treated with E2 also showed increases in the amplitude of pH-sensitive potassium currents, as assessed by whole-cell recordings. These currents are blocked by clofilium. Although confocal microscopy suggested that most of the channels are located in intracellular compartments, the increase in macroscopic currents suggests that E2 treatment increases the number of active channels at the cell surface. Application of small interfering RNA specific for KCNK5 decreased pH-sensitive potassium currents and also reduced the estrogen-induced proliferation of T47D cells. We conclude that E2 induces the expression of KCNK5 via ERα(+) in breast cancer cells, and this channel plays a role in regulating proliferation in these cell lines. KCNK5 may therefore represent a useful target for treatment, for example, of tamoxifen-resistant breast cancer.
Collapse
Affiliation(s)
- Claudia P Alvarez-Baron
- Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204-5001, USA
| | | | | | | | | |
Collapse
|
64
|
Kunzelmann K, Kongsuphol P, Chootip K, Toledo C, Martins JR, Almaca J, Tian Y, Witzgall R, Ousingsawat J, Schreiber R. Role of the Ca2+-activated Cl- channels bestrophin and anoctamin in epithelial cells. Biol Chem 2011; 392:125-34. [DOI: 10.1515/bc.2011.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Two families of proteins, the bestrophins (Best) and the recently cloned TMEM16 proteins (anoctamin, Ano), recapitulate properties of Ca2+-activated Cl- currents. Best1 is strongly expressed in the retinal pigment epithelium and could have a function as a Ca2+-activated Cl- channel as well as a regulator of Ca2+ signaling. It is also present at much lower levels in other cell types including epithelial cells, where it regulates plasma membrane localized Cl- channels by controlling intracellular Ca2+ levels. Best1 interacts with important Ca2+-signaling proteins such as STIM1 and can interact directly with other Ca2+-activated Cl- channels such as TMEM16A. Best1 is detected in the endoplasmic reticulum (ER) where it shapes the dynamic ER structure and regulates cell proliferation, which could be important for renal cystogenesis. Ca2+-activated Cl- channels of the anoctamin family (TMEM16A) show biophysical and pharmacological properties that are typical for endogenous Ca2+-dependent Cl- channels. TMEM16 proteins are abundantly expressed and many reports demonstrate their physiological importance in epithelial as well as non-epithelial cells. These channels are also activated by cell swelling and can therefore control cell volume, proliferation and apoptosis. To fully understand the function and regulation of Ca2+-activated Cl- currents, it is necessary to appreciate that Best1 and TMEM16A are embedded in a protein network and that they probably operate in functional microdomains.
Collapse
|
65
|
Park HA, Jung SY, Lee SH, Kang HB, Min MS, Kim JA, Choo DJ, Oh CR, Kim YD, Lee KT, Lee JY. Anti-Cancer Activity of T-Type Calcium Channel Blocker In Vivo. B KOREAN CHEM SOC 2010. [DOI: 10.5012/bkcs.2010.31.11.3353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
66
|
Restrepo-Angulo I, De Vizcaya-Ruiz A, Camacho J. Ion channels in toxicology. J Appl Toxicol 2010; 30:497-512. [DOI: 10.1002/jat.1556] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
67
|
Wang QJ, Cai XB, Liu MH, Hu H, Tan XJ, Jing XB. Apoptosis induced by emodin is associated with alterations of intracellular acidification and reactive oxygen species in EC-109 cells. Biochem Cell Biol 2010; 88:767-74. [PMID: 20651850 DOI: 10.1139/o10-020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Emodin (1,3,8-trihydroxy-6-methylanthraquinone), a natural anthraquinone derivative found in several herbal medicines, is highly active in suppressing the proliferation of various tumor cells such as breast, hepatocellular, and lung cancer cells under in vitro conditions. The mechanism of emodin-induced apoptosis in esophagus carcinoma cells, EC-109, is not completely understood. In this study, EC-109 cells treated with emodin underwent rapid apoptosis as judged by morphological changes and flow cytometry analysis. The addition of emodin to EC-109 cells led to the inhibition of growth in a time- and dose-dependent manner. Fluorescence measurements of cells indicated that the intracellular pH (pHi) decreased significantly by 0.47-0.78 units. The results obtained from flow cytometry suggested that bursts of reactive oxygen species took place after the application of emodin. The present study indicates that emodin may be a strong anticancer drug against esophagus cancer cells by causing various early events leading to growth inhibition, including the production of reactive oxygen species and decrease of pHi, which may result in cellular apoptosis.
Collapse
Affiliation(s)
- Q J Wang
- Department of Gastroenterology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong Province, P.R. China
| | | | | | | | | | | |
Collapse
|
68
|
Xu B, Mao J, Wang L, Zhu L, Li H, Wang W, Jin X, Zhu J, Chen L. ClC-3 chloride channels are essential for cell proliferation and cell cycle progression in nasopharyngeal carcinoma cells. Acta Biochim Biophys Sin (Shanghai) 2010; 42:370-80. [PMID: 20539936 DOI: 10.1093/abbs/gmq031] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
ClC-3, a gene encoding a candidate protein for volume-activated chloride (C(-)) channels, may be involved in tumor development. Herein we report a study using an antisense "knock-down" strategy to investigate the mechanism by which ClC-3 affects cell proliferation in nasopharyngeal carcinoma CNE-2Z cells. With immunoblots and MTT assays we demonstrated that the expression of ClC-3 was cell cycle dependent and in a similar concentration-dependent manner, an antisense oligonucleotide specific for ClC-3 inhibited ClC-3 protein expression and cell proliferation. The expression level of ClC-3 correlated with cell proliferation. Moreover, in the cells exposed to a ClC-3 antisense oligonucleotide, the cloning efficiency was inhibited, and cells were arrested in the S phase. The ClC-3 antisense oligonucleotide inhibited the volume-activated C(-) current (I(Cl,vol)) and the regulatory volume decrease (RVD) in a concentration-dependent manner. Additionally, the I(Cl,vol) or RVD was positively correlated with cell proliferation in the treated cells. In conclusion, ClC-3 is involved in cell proliferation and cell cycle progression through a mechanism involving modulation of I(Cl,vol) and RVD. CIC-3 may represent a therapeutic target in human cancer.
Collapse
Affiliation(s)
- Bin Xu
- Guangdong Pharmaceutical University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Yasuda T, Adams DJ. Physiological roles of ion channels in adult neural stem cells and their progeny. J Neurochem 2010; 114:946-59. [PMID: 20492359 DOI: 10.1111/j.1471-4159.2010.06822.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Elucidation of the machinery of adult neurogenesis is indispensable for the treatment of neurodegenerative diseases by therapeutic drugs and/or by neural stem cell (NSC) transplantation. It is well known that membrane ion channels play a critical role in cell function, including proliferation, apoptosis and migration in a wide range of cells. In NSC research, interdisciplinary collaboration between cell biologists and membrane physiologists has been pursued principally to monitor ion channel and synaptic currents as a hallmark of neuronal differentiation and maturation of NSC progeny. Nevertheless, less attention had been paid to a functional role of ion channels in NSCs or their immature progeny. Recently, however, evidence regarding their functional relevance has started to accumulate. In focusing on the early stages of the neurogenic process during which NSCs give rise to neuroblasts, this review highlights the latent ability of ion channels to act as functional regulators of adult neurogenesis.
Collapse
Affiliation(s)
- Takahiro Yasuda
- Health Innovations Research Institute, RMIT University, Melbourne, Victoria, Australia.
| | | |
Collapse
|
70
|
Hedberg D, Wallin M. Effects of Roundup and glyphosate formulations on intracellular transport, microtubules and actin filaments in Xenopus laevis melanophores. Toxicol In Vitro 2010; 24:795-802. [PMID: 20036731 DOI: 10.1016/j.tiv.2009.12.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 11/17/2009] [Accepted: 12/22/2009] [Indexed: 11/25/2022]
Abstract
Glyphosate containing herbicides, such as Roundup, are commonly used and generally considered to be safe. However, some toxic effects are found on amphibians in vivo and human and mouse cells in vitro. In this study the effects of Roundup, glyphosate, glyphosateisopropylamine and isopropylamine were studied on intracellular transport by measuring aggregation capacity in Xenopus laevis melanophores. The chemicals inhibited retrograde transport of melanosomes in the range of 0.5-5mM. Cellular morphology and localization of microtubules and actin filaments were affected as determined by immunocytochemistry. Both glyphosate and Roundup decreased pH in the media. Acidic pH inhibited melanosome transport and altered microtubule and actin morphology in the absence of chemicals, while transport inhibiting concentrations of glyphosate, Roundup and glyphosateisopropylamine disassembled both microtubules and actin filaments. At physiological pH the effects of Roundup decreased whereas glyphosate failed to inhibit transport. Physiological pH decreases glyphosate lipophilicity and its diffusion into the cytoplasm. The Roundup formulation contains surfactants, such as POEA (polyetylated tallow amine) that increases membrane permeability allowing cellular uptake at physiological pH. Our results show that the effects of glyphosate containing compounds are pH-dependent and that they inhibit intracellular transport through disassembly of the cytoskeleton possibly by interfering with intracellular Ca(2+)-balance.
Collapse
Affiliation(s)
- Daniel Hedberg
- Department of Zoology, University of Gothenburg, PO Box 463, SE-405 30 Göteborg, Sweden
| | | |
Collapse
|
71
|
Rapamycin inhibits oncogenic intestinal ion channels and neoplasia in APC(Min/+) mice. Oncogene 2009; 29:1553-60. [PMID: 19966863 DOI: 10.1038/onc.2009.435] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The adenomatous polyposis coli (APC) gene is mutated in familial adenomatous polyposis. Mice with a heterozygous APC(Min) mutation develop multiple intestinal neoplasia (Min) leading to premature death. Early in colorectal carcinogenesis, APC(Min/+) mice show enhanced Akt-mammalian target of rapamycin (mTOR) signaling, which is paralleled by upregulation of oncogenic K(+) channels. In this study, we tested the effect of mTOR inhibition with rapamycin on tumor formation in APC(Min/+) mice and evaluated ion channel regulation. We found that continuous long-term rapamycin treatment of APC(Min/+) mice dramatically inhibits intestinal neoplasia. Moreover, although untreated APC(Min/+) mice lose weight, experience intestinal bleeding and succumb to multiple neoplasia by 22.3+/-1.4 weeks of age, mice treated with rapamycin maintain stable weight and survive long term (39.6+/-3.4 weeks), with more than 30% surviving >1 year. Impressively, abnormalities in colonic electrolyte transport typical for APC(Min/+) mice are abolished, along with the suppression of epithelial Na(+) channel (ENaC) and oncogenic K(+) ion channels BK, Elk1 and Erg1, both functionally and at mRNA levels. These results show that continuous prophylaxis by rapamycin markedly inhibits the development of APC mutation-related polyposis, and suggest a novel contributing mechanism of action through the blockade of intestinal oncogenic ion channels.
Collapse
|
72
|
Barro-Soria R, Aldehni F, Almaça J, Witzgall R, Schreiber R, Kunzelmann K. ER-localized bestrophin 1 activates Ca2+-dependent ion channels TMEM16A and SK4 possibly by acting as a counterion channel. Pflugers Arch 2009; 459:485-97. [PMID: 19823864 DOI: 10.1007/s00424-009-0745-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 09/29/2009] [Accepted: 09/30/2009] [Indexed: 01/14/2023]
Abstract
Bestrophins form Ca(2+)-activated Cl(-) channels and regulate intracellular Ca(2+) signaling. We demonstrate that bestrophin 1 is localized in the endoplasmic reticulum (ER), where it interacts with stromal interacting molecule 1, the ER-Ca(2+) sensor. Intracellular Ca(2+) transients elicited by stimulation of purinergic P2Y(2) receptors in HEK293 cells were augmented by hBest1. The p21-activated protein kinase Pak2 was found to phosphorylate hBest1, thereby enhancing Ca(2+) signaling and activation of Ca(2+)-dependent Cl(-) (TMEM16A) and K(+) (SK4) channels. Lack of bestrophin 1 expression in respiratory epithelial cells of mBest1 knockout mice caused expansion of ER cisterns and induced Ca(2+) deposits. hBest1 is, therefore, important for Ca(2+) handling of the ER store and may resemble the long-suspected counterion channel to balance transient membrane potentials occurring through inositol triphosphate (IP(3))-induced Ca(2+) release and store refill. Thus, bestrophin 1 regulates compartmentalized Ca(2+) signaling that plays an essential role in Best macular dystrophy, inflammatory diseases such as cystic fibrosis, as well as proliferation.
Collapse
Affiliation(s)
- René Barro-Soria
- Institut für Physiologie, Universität Regensburg, Universitätsstrasse 31, 93053 Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
73
|
Bestrophin and TMEM16-Ca(2+) activated Cl(-) channels with different functions. Cell Calcium 2009; 46:233-41. [PMID: 19783045 DOI: 10.1016/j.ceca.2009.09.003] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 09/01/2009] [Accepted: 09/04/2009] [Indexed: 12/23/2022]
Abstract
In the past, a number of candidates have been proposed to form Ca(2+) activated Cl(-) currents, but it is only recently that two families of proteins, the bestrophins and the TMEM16-proteins, recapitulate reliably the properties of Ca(2+) activated Cl(-) currents. Bestrophin 1 is strongly expressed in the retinal pigment epithelium, but also at lower levels in other cell types. Bestrophin 1 may form Ca(2+) activated chloride channels and, at the same time, affect intracellular Ca(2+) signaling. In epithelial cells, bestrophin 1 probably controls receptor mediated Ca(2+) signaling. It may do so by facilitating Ca(2+) release from the endoplasmic reticulum, thereby indirectly activating membrane localized Ca(2+)-dependent Cl(-) channels. In contrast to bestrophin 1, the Ca(2+) activated Cl(-) channel TMEM16A (anoctamin 1, ANO1) shows most of the biophysical and pharmacological properties that have been attributed to Ca(2+)-dependent Cl(-) channels in various tissues. TMEM16A is broadly expressed in both mouse and human tissues and is of particular importance in epithelial cells. Thus exocrine gland secretion as well as electrolyte transport by both respiratory and intestinal epithelia requires TMEM16A. Because of its role for Ca(2+)-dependent Cl(-) secretion in human airways, it is likely to become a prime target for the therapy of cystic fibrosis lung disease, caused by defective cAMP-dependent Cl(-) secretion. It will be very exciting to learn, how TMEM16A and other TMEM16-proteins are activated upon increase in intracellular Ca(2+), and whether the other nine members of the TMEM16 family also form Cl(-) channels with properties similar to TMEM16A.
Collapse
|
74
|
Klimmeck D, Daiber PC, Brühl A, Baumann A, Frings S, Möhrlen F. Bestrophin 2: an anion channel associated with neurogenesis in chemosensory systems. J Comp Neurol 2009; 515:585-99. [PMID: 19480000 DOI: 10.1002/cne.22075] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The chemosensory neuroepithelia of the vertebrate olfactory system share a life-long ability to regenerate. Novel neurons proliferate from basal stem cells that continuously replace old or damaged sensory neurons. The sensory neurons of the mouse and rat olfactory system specifically express bestrophin 2, a member of the bestrophin family of calcium-activated chloride channels. This channel was recently proposed to operate as a transduction channel in olfactory sensory cilia. We raised a polyclonal antibody against bestrophin 2 and characterized the expression pattern of this protein in the mouse main olfactory epithelium, septal organ of Masera, and vomeronasal organ. Comparison with the maturation markers growth-associated protein 43 and olfactory marker protein revealed that bestrophin 2 was expressed in developing sensory neurons of all chemosensory neuroepithelia, but was restricted to proximal cilia in mature sensory neurons. Our results suggest that bestrophin 2 plays a critical role during differentiation and growth of axons and cilia. In mature olfactory receptor neurons, it appears to support growth and function of sensory cilia.
Collapse
Affiliation(s)
- Daniel Klimmeck
- Department of Molecular Physiology, Institute of Zoology, University of Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
75
|
Differentiation impairs low pH-induced Ca2+ signaling and ERK phosphorylation in granule precursor tumour cells. Cell Calcium 2009; 45:391-9. [PMID: 19249096 DOI: 10.1016/j.ceca.2009.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 12/18/2008] [Accepted: 01/15/2009] [Indexed: 12/19/2022]
Abstract
Extracellular acidification is a hallmark of a number of debilitating pathologies including cancer, ischemia and inflammation. We have recently shown that in human granule precursor tumour cells a fall in extracellular pH triggers increases in intracellular Ca(2+) concentration through activation of G-protein coupled proton-sensing receptors coupling to phospholipase C. This pH-dependent rise in cytosolic Ca(2+) led to activation of the extracellular signal-regulated kinase ERK, providing a mechanistic explanation of how extracellular acidification can promote tumour growth. We now find that differentiation of granule precursor tumour cells profoundly affects their ability to respond to extracellular acidification with gene transcription. Differentiating cells have a lower Ca(2+) release probability from intracellular Ca(2+) stores upon acidification and cells that respond have a significantly smaller and slower Ca(2+) signal than proliferating cells. Importantly, Ca(2+) release in differentiating cells fails to evoke ERK phosphorylation. This altered responsiveness of differentiating cells is not due to reduced proton-sensing receptor expression or diminished Ca(2+) store content. Rather, our results suggest that in differentiating cells, the proton-sensing receptor couples less effectively to phospholipase C activation and IP(3) formation. Hence, the ability of human granule cells to respond to extracellular acidification by generating Ca(2+) signals and ERK activation is state-dependent, being lost upon differentiation.
Collapse
|
76
|
The manipulation of calcium oscillations by harnessing self-organisation. Biosystems 2008; 94:153-63. [PMID: 18606209 DOI: 10.1016/j.biosystems.2008.05.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Revised: 10/29/2007] [Accepted: 05/23/2008] [Indexed: 11/21/2022]
|
77
|
Huang WC, Swietach P, Vaughan-Jones RD, Ansorge O, Glitsch MD. Extracellular acidification elicits spatially and temporally distinct Ca2+ signals. Curr Biol 2008; 18:781-785. [PMID: 18485712 DOI: 10.1016/j.cub.2008.04.049] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Revised: 04/08/2008] [Accepted: 04/21/2008] [Indexed: 11/29/2022]
Abstract
Extracellular acidification accompanies neoplastic transformation of tissues and increases with tumor aggressiveness [1, 2]. The intracellular signaling cascade triggered by this process remains poorly understood and may be linked to recently discovered proton-activated G protein-coupled receptors such as OGR1 and G2A [3, 4]. Here, we report that OGR1 and G2A are expressed in human medulloblastoma tissue and its corresponding neuronal cell line. We show that extracellular acidification activates phospholipase C, IP(3) formation, and subsequent Ca2+ release from thapsigargin-sensitive stores in neurons. The number of responsive cells and the amount of Ca2+ released from stores correlated positively with the extent of extracellular acidification. Ca2+ release recruited the MEK/ERK pathway, providing a mechanistic explanation for how acidification stimulates cell growth. In addition, acidification activated Ca2+-permeable ion channels through a mechanism dependent on phospholipase C but independent of store depletion or a cytoplasmic Ca2+ rise. Hence, extracellular acidification, to levels seen in tumor tissue, activates temporally and spatially distinct pathways that elevate Ca2+ and may be directly relevant for tumor cell biology.
Collapse
Affiliation(s)
- Wan-Chen Huang
- Department of Physiology, Anatomy, and Genetics, Sherrington Building, Oxford University, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Pawel Swietach
- Proton Transport Group, Sherrington Building, Oxford University, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Richard D Vaughan-Jones
- Proton Transport Group, Sherrington Building, Oxford University, Parks Road, Oxford OX1 3PT, United Kingdom
| | - Olaf Ansorge
- Department of Neuropathology, John Radcliffe Hospital, Headley Way, Headington, Oxford OX3 9DU, United Kingdom
| | - Maike D Glitsch
- Department of Physiology, Anatomy, and Genetics, Sherrington Building, Oxford University, Parks Road, Oxford OX1 3PT, United Kingdom.
| |
Collapse
|
78
|
Franco R, Panayiotidis MI, de la Paz LDO. Autocrine signaling involved in cell volume regulation: the role of released transmitters and plasma membrane receptors. J Cell Physiol 2008; 216:14-28. [PMID: 18300263 DOI: 10.1002/jcp.21406] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cell volume regulation is a basic homeostatic mechanism transcendental for the normal physiology and function of cells. It is mediated principally by the activation of osmolyte transport pathways that result in net changes in solute concentration that counteract cell volume challenges in its constancy. This process has been described to be regulated by a complex assortment of intracellular signal transduction cascades. Recently, several studies have demonstrated that alterations in cell volume induce the release of a wide variety of transmitters including hormones, ATP and neurotransmitters, which have been proposed to act as extracellular signals that regulate the activation of cell volume regulatory mechanisms. In addition, changes in cell volume have also been reported to activate plasma membrane receptors (including tyrosine kinase receptors, G-protein coupled receptors and integrins) that have been demonstrated to participate in the regulatory process of cell volume. In this review, we summarize recent studies about the role of changes in cell volume in the regulation of transmitter release as well as in the activation of plasma membrane receptors and their further implications in the regulation of the signaling machinery that regulates the activation of osmolyte flux pathways. We propose that the autocrine regulation of Ca2+-dependent and tyrosine phosphorylation-dependent signaling pathways by the activation of plasma membrane receptors and swelling-induced transmitter release is necessary for the activation/regulation of osmolyte efflux pathways and cell volume recovery. Furthermore, we emphasize the importance of studying these extrinsic signals because of their significance in the understanding of the physiology of cell volume regulation and its role in cell biology in vivo, where the constraint of the extracellular space might enhance the autocrine or even paracrine signaling induced by these released transmitters.
Collapse
Affiliation(s)
- Rodrigo Franco
- Laboratory of Cell Biology and Signal Transduction, Biomedical Research Unit, FES-Iztacala, UNAM, Mexico.
| | | | | |
Collapse
|
79
|
Taylor JT, Huang L, Pottle JE, Liu K, Yang Y, Zeng X, Keyser BM, Agrawal KC, Hansen JB, Li M. Selective blockade of T-type Ca2+ channels suppresses human breast cancer cell proliferation. Cancer Lett 2008; 267:116-24. [PMID: 18455293 DOI: 10.1016/j.canlet.2008.03.032] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Revised: 03/04/2008] [Accepted: 03/10/2008] [Indexed: 12/14/2022]
Abstract
We have measured the expression of T-type Ca2+ channel mRNA in breast cancer cell lines (MCF-7 (ERalpha+) using Western blot and quantitative real-time PCR (Q-RT-PCR). These results revealed that the MCF-7 cells express both alpha1G and alpha1H isoforms of T-type Ca2+ channels. In order to further clarify the role of T-type Ca2+ channels in proliferation, we tested the effects of a selective T-type Ca2+ channel inhibitor NNC-55-0396 on cellular proliferation. MCF-7 (ERalpha+) cellular proliferation was inhibited by the compound. In contrast, NNC-55-0396 at same concentration had no effect on the proliferation of MCF-10A cells, a non-cancer breast epithelial cell line. We also found that message expression of the T-type Ca2+ channels were only expressed in rapidly growing non-confluent cells but not in the cytostatic confluent cells. Knocking down the expression of T-type Ca2+ channels with siRNA targeting both alpha1G and alpha1H resulted in growth inhibition as much as 45%+/-5.0 in MCF-7 cells as compared to controls. In conclusion, our results suggest that T-type Ca2+ channel antagonism/silencing may reduce cellular proliferation in mitogenic breast cells.
Collapse
Affiliation(s)
- James T Taylor
- Department of Pharmacology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Proliferation of human lens epithelial cells (HLE-B3) is inhibited by blocking of voltage-gated calcium channels. Pflugers Arch 2008; 457:47-59. [DOI: 10.1007/s00424-008-0514-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Revised: 03/19/2008] [Accepted: 04/04/2008] [Indexed: 10/22/2022]
|
81
|
Nakamura K, Kamouchi M, Kitazono T, Kuroda J, Matsuo R, Hagiwara N, Ishikawa E, Ooboshi H, Ibayashi S, Iida M. Role of NHE1 in calcium signaling and cell proliferation in human CNS pericytes. Am J Physiol Heart Circ Physiol 2008; 294:H1700-7. [PMID: 18263712 DOI: 10.1152/ajpheart.01203.2007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The central nervous system (CNS) pericytes play an important role in brain microcirculation. Na(+)/H(+) exchanger isoform 1 (NHE1) has been suggested to regulate the proliferation of nonvascular cells through the regulation of intracellular pH, Na(+), and cell volume; however, the relationship between NHE1 and intracellular Ca(2+), an essential signal of cell growth, is still not known. The aim of the present study was to elucidate the role of NHE1 in Ca(2+) signaling and the proliferation of human CNS pericytes. The intracellular Ca(2+) concentration was measured by fura 2 in cultured human CNS pericytes. The cells showed spontaneous Ca(2+) oscillation under quasi-physiological ionic conditions. A decrease in extracellular pH or Na(+) evoked a transient Ca(2+) rise followed by Ca(2+) oscillation, whereas an increase in pH or Na(+) did not induce the Ca(2+) responses. The Ca(2+) oscillation was inhibited by an inhibitor of NHE in a dose-dependent manner and by knockdown of NHE1 by using RNA interference. The Ca(2+) oscillation was completely abolished by thapsigargin. The proliferation of pericytes was attenuated by inhibition of NHE1. These results demonstrate that NHE1 regulates Ca(2+) signaling via the modulation of Ca(2+) release from the endoplasmic reticulum, thus contributing to the regulation of proliferation in CNS pericytes.
Collapse
Affiliation(s)
- Kuniyuki Nakamura
- Dept. of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Ousingsawat J, Spitzner M, Schreiber R, Kunzelmann K. Upregulation of colonic ion channels in APC ( Min/+ ) mice. Pflugers Arch 2008; 456:847-55. [PMID: 18247045 DOI: 10.1007/s00424-008-0451-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Revised: 11/21/2007] [Accepted: 01/09/2008] [Indexed: 12/16/2022]
Abstract
The adenomatous polyposis coli (APC) tumor suppressor gene is mutated in almost all human colonic cancers. Disturbances in Na(+) absorption have been observed in colonic cancer, and ion channels such as ether a go-go (Eag) or Ca(2+)-sensitive BK channels have been recognized for their oncogenic potential. APC ( Min/+ ) mice have reduced APC expression and develop multiple intestinal neoplasias (Min). Ion channels in the colonic epithelium were examined using electrophysiology and molecular techniques. APC ( Min/+ ) mice developed intestinal neoplasia and experienced a significant weight loss. Due to intestinal bleedings, the hematocrit was largely reduced and plasma aldosterone levels were enhanced. Rectal potential measurements in vivo indicated an increase in amiloride-sensitive Na(+) absorption in APC ( Min/+ ) mice. Quantitative Ussing chamber studies demonstrated enhanced Na(+) absorption via epithelial Na(+) channels (ENaC) and suggested enhanced activity of oncogenic BK and Eag-1 channels. Patch clamp and fluorescence measurements on isolated crypts suggested enhanced K(+) channel activity in the surface epithelium. ENaC-mRNA and membrane protein expression was enhanced in colonic surface epithelial cells. The data suggest that reduced expression of the APC gene with upregulation of the downstream proteins Akt and mTOR and subsequent hyperaldosteronism is paralleled by upregulation of oncogenic potassium channels and enhanced colonic Na(+) absorption.
Collapse
Affiliation(s)
- Jiraporn Ousingsawat
- Institut für Physiologie, Universität Regensburg, Universitätsstrasse 31, 93053, Regensburg, Germany
| | | | | | | |
Collapse
|
83
|
Abstract
Survival of human and animal cells requires avoidance of excessive alterations of cell volume. The osmolarity amassed by cellular accumulation of organic substances must be compensated by lowering cytosolic ion concentrations. The Na+/K+ ATPase extrudes Na+ in exchange for K+, which can permeate the cell membrane through K+ channels. K+ exit generates a cell-negative potential difference across the cell membrane, driving the exit of anions such as Cl-. The low cytosolic Cl- concentrations counterbalance the excess cellular osmolarity by organic substances. Cell volume regulation following cell swelling involves releasing ions through activation of K+ channels and/or anion channels, KCl-cotransport, or parallel activation of K+/H+ exchange and Cl-/HCO3- exchange. Cell volume regulation following cell shrinkage involves accumulation of ions through activation of Na+,K+,2Cl- cotransport, Na+/H+ exchange in parallel to Cl-/HCO3- exchange, or Na+ channels. The Na+ taken up is extruded by the Na+/K+ ATPase in exchange for K+. Shrunken cells further accumulate organic osmolytes such as sorbitol and glycerophosphorylcholine, and monomeric amino acids by altered metabolism and myoinositol (inositol), betaine, taurine, and amino acids by Na+ coupled transport. They release osmolytes during cell swelling. Challenges of cell volume homeostasis include transport, hormones, transmitters, and drugs. Moreover, alterations of cell volume participate in the machinery regulating cell proliferation and apoptotic cell death. Deranged cell volume regulation significantly contributes to the pathophysiology of several disorders such as liver insufficiency, diabetic ketoacidosis, hypercatabolism, fibrosing disease, sickle cell anemia, and infection.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology I, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
84
|
Abstract
The inwardly rectifying potassium channel Kir4.1 is widely expressed by astrocytes throughout the brain. Kir4.1 channels are absent in immature, proliferating glial cells. The progressive expression of Kir4.1 correlates with astrocyte differentiation and is characterized by the establishment of a negative membrane potential (> -70 mV) and an exit from the cell cycle. Despite some correlative evidence, a mechanistic interdependence between Kir4.1 expression, membrane hyperpolarization, and control of cell proliferation has not been demonstrated. To address this question, we used astrocyte-derived tumors (glioma) that lack functional Kir4.1 channels, and generated two glioma cell lines that stably express either AcGFP-tagged Kir4.1 channels or AcGFP vectors only. Kir4.1 expression confers the same K+ conductance to glioma membranes and a similar responsiveness to changes in [K+]o that characterizes differentiated astrocytes. Kir4.1 expression was sufficient to move the resting potential of gliomas from -50 to -80 mV. Importantly, Kir4.1 expression impaired cell growth by shifting a significant number of cells from the G2/M phase into the quiescent G0/G1 stage of the cell cycle. Furthermore, these effects could be nullified entirely if Kir4.1 channels were either pharmacologically inhibited by 100 microM BaCl2 or if cells were chronically depolarized by 20 mM KCl to the membrane voltage of growth competent glioma cells. These studies therefore demonstrate directly that Kir4.1 causes a membrane hyperpolarization that is sufficient to account for the growth attenuation, which in turn induces cell maturation characterized by a shift of the cells from G2/M into G0/G1.
Collapse
Affiliation(s)
- HARUKI HIGASHIMORI
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
- Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - HARALD SONTHEIMER
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
- Center for Glial Biology in Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
85
|
Obara M, Szeliga M, Albrecht J. Regulation of pH in the mammalian central nervous system under normal and pathological conditions: facts and hypotheses. Neurochem Int 2007; 52:905-19. [PMID: 18061308 DOI: 10.1016/j.neuint.2007.10.015] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Revised: 10/17/2007] [Accepted: 10/22/2007] [Indexed: 11/27/2022]
Abstract
The maintenance of pH homeostasis in the CNS is of key importance for proper execution and regulation of neurotransmission, and deviations from this homeostasis are a crucial factor in the mechanism underlying a spectrum of pathological conditions. The first few sections of the review are devoted to the brain operating under normal conditions. The article commences with an overview of how extrinsic factors modelling the brain at work: neurotransmitters, depolarising stimuli (potassium and voltage changes) and cyclic nucleotides as major signal transducing vehicles affect pH in the CNS. Further, consequences of pH alterations on the major aspects of CNS function and metabolism are outlined. Next, the major cellular events involved in the transport, sequestration, metabolic production and buffering of protons that are common to all the mammalian cells, including the CNS cells. Since CNS function reflects tight interaction between astrocytes and neurons, the pH regulatory events pertinent to either cell type are discussed: overwhelming evidence implicates astrocytes as a key player in pH homeostasis in the brain. The different classes of membrane proteins involved in proton shuttling are listed and their mechanisms of action are given. These include: the Na+/H+ exchanger, different classes of bicarbonate transporters acting in a sodium-dependent- or -independent mode, monocarboxylic acid transporters and the vacuolar-type proton ATPase. A separate section is devoted to carbonic anhydrase, which is represented by multiple isoenzymes capable of pH buffering both in the cell interior and in the extracellular space. Next, impairment of pH regulation and compensatory responses occurring in brain affected by different pathologies: hypoxia/ischemia, epilepsy, hyperammonemic encephalopathies, cerebral tumours and HIV will be described. The review is limited to facts and plausible hypotheses pertaining to phenomena directly involved in pH regulation: changes in pH that accompany metabolic stress but have no distinct implications for the pH regulatory mechanisms are not dealt with. In most cases, the vast body of knowledge derived from in vitro studies remains to be verified in in vivo settings.
Collapse
Affiliation(s)
- Marta Obara
- Department of Neurotoxicology, Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Street, 02-106 Warsaw, Poland
| | | | | |
Collapse
|
86
|
Bjerregaard H. Effects of cadmium on differentiation and cell cycle progression in cultured Xenopus kidney distal epithelial (A6) cells. Altern Lab Anim 2007; 35:343-8. [PMID: 17650953 DOI: 10.1177/026119290703500302] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cadmium (Cd) is an important industrial and environmental pollutant, and the kidney is the primary organ to be affected. To elucidate the effects of Cd on cell proliferation, an epithelial cell line (A6) originally derived from the distal part of the Xenopus laevis kidney was cultured in media containing 10% fetal bovine serum. The effects of Cd (added as CdCl(2)) on cellular growth and differentiation from single cells to confluent epithelia were investigated by visual inspection and by measurement of the degree to which living cells covered a unit area. Over a concentration range from 5 to 50 microM, Cd did not affect the settling and adherence of single cells to the bottom of the culture well. The addition of 5 microM Cd for 4 days did not affect the ability of the A6 cells to develop confluent epithelia, measured as the area covered by adherent living epithelial cells (99 +/- 4% of the control value). However, 10 microM Cd did effectively inhibit development of confluent epithelia to 13 +/- 5% compared to control. Visual inspection of adherent cells exposed to 50 microM Cd for 7 days revealed no increase in cell number or in cell death, which indicated the induction of cell cycle arrest. Flow cytometric analysis showed that treatment of cells with Cd (0.4mM) for 24 hours induced a significant increase in the proportion of G1 phase cells from 58.6 +/- 3.9 to 80.6 +/- 3.7%, and a corresponding reduction in the proportion of cells in both the S and G2 phases from 24.0 +/- 3.6 to 13.4 +/- 3.3% and 17.2 +/- 1.7 to 5.8 +/- 2.1%, respectively. This study showed that Cd stopped cell proliferation in a very narrow concentration range, between 5 and 10 microM, and cell cycle analysis indicated that Cd arrested the cells in the G1 phase of the cell cycle.
Collapse
Affiliation(s)
- Henning Bjerregaard
- Department of Life Sciences and Chemistry, Roskilde University, Roskilde, Denmark.
| |
Collapse
|
87
|
Ding XW, Luo HS. Ether à go-go potassium channel and malignant tumors. Shijie Huaren Xiaohua Zazhi 2007; 15:2772-2779. [DOI: 10.11569/wcjd.v15.i26.2772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recently, increasing evidence from cell biology and pharmacology demonstrates that cancer cells exhibit ion channel expression patterns, ion conductances and electric properties that are very different from those of resting cells. These peculiar properties are functionally involved in cancer pathogenesis. In particular, because of its oncogenic properties, distribution, modulation and pharmacology, human ether à go-go potassium channel (Eag1, Kv10.1, KCNH1) is considered a critical ion channel-encoding gene involved in the establishment and maintenance of neoplastic growth. This review summarizes most of the findings regarding Eag1 channels and malignant tumors, focusing on cellular mechanisms, mRNA and protein expression in tissues, oncogenic properties, modulation and pharmacology.
Collapse
|
88
|
Martínez D, Vermeulen M, von Euw E, Sabatté J, Maggíni J, Ceballos A, Trevani A, Nahmod K, Salamone G, Barrio M, Giordano M, Amigorena S, Geffner J. Extracellular acidosis triggers the maturation of human dendritic cells and the production of IL-12. THE JOURNAL OF IMMUNOLOGY 2007; 179:1950-9. [PMID: 17641062 DOI: 10.4049/jimmunol.179.3.1950] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although the development of an acidic tissue environment or acidosis is a hallmark of inflammatory processes, few studies analyze the effect of extracellular pH on immune cells. We have previously shown that exposure of murine dendritic cells (DCs) to pH 6.5 stimulates macropinocytosis and cross-presentation of extracellular Ags by MHC class I molecules. We report that the transient exposure of human DCs to pH 6.5 markedly increases the expression of HLA-DR, CD40, CD80, CD86, CD83, and CCR7 and improves the T cell priming ability of DCs. Incubation of DCs at pH 6.5 results in the activation of the PI3K/Akt and the MAPK pathways. Using specific inhibitors, we show that the maturation of DCs induced by acidosis was strictly dependent on the activation of p38 MAPK. DC exposure to pH 6.5 also induces a dramatic increase in their production of IL-12, stimulating the synthesis of IFN-gamma, but not IL-4, by Ag-specific CD4(+) T cells. Interestingly, we find that suboptimal doses of LPS abrogated the ability of pH 6.5 to induce DC maturation, suggesting a cross-talk between the activation pathways triggered by LPS and extracellular protons in DCs. We conclude that extracellular acidosis in peripheral tissues may contribute to the initiation of adaptive immune responses by DCs, favoring the development of Th1 immunity.
Collapse
Affiliation(s)
- Diego Martínez
- Institute of Hematologic Research, National Academy of Medicine, National Reference Center for AIDS, and Department of Microbiology, Buenos Aires University School of Medicine, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Ousingsawat J, Spitzner M, Puntheeranurak S, Terracciano L, Tornillo L, Bubendorf L, Kunzelmann K, Schreiber R. Expression of voltage-gated potassium channels in human and mouse colonic carcinoma. Clin Cancer Res 2007; 13:824-31. [PMID: 17289873 DOI: 10.1158/1078-0432.ccr-06-1940] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE Voltage-gated Kv potassium channels, like ether a go-go (EAG) channels, have been recognized for their oncogenic potential in breast cancer and other malignant tumors. EXPERIMENTAL DESIGN We examined the molecular and functional expression of Kv channels in human colonic cancers and colon of mice treated with the chemical carcinogens dimethylhydrazine and N-methyl-N-nitrosourea. The data were compared with results from control mice and animals with chemically induced DSS colitis. RESULTS Electrogenic salt transport by amiloride-sensitive Na+ channels and cyclic AMP-activated cystic fibrosis transmembrane conductance regulator Cl- channels were attenuated during tumor development and colitis, whereas Ca2+-dependent transport remained unchanged. Kv channels, in particular Eag-1, were enhanced during carcinogenesis. Multiplex reverse transcription-PCR showed increased mRNA expression for Kv1.3, Kv1.5, Kv3.1, and members of the Eag channel family, after dimethylhydrazine and N-methyl-N-nitrosourea treatment. Eag-1 protein was detected in the malignant mouse colon and human colonic cancers. Genomic amplification of Eag-1 was found in 3.4% of all human colorectal adenocarcinoma and was an independent marker of adverse prognosis. CONCLUSIONS The study predicts an oncogenic role of Kv and Eag channels for the development of colonic cancer. These channels may represent an important target for a novel pharmacotherapy of colonic cancer.
Collapse
Affiliation(s)
- Jiraporn Ousingsawat
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany, and Institute for Pathology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Pan Z, Capó-Aponte JE, Zhang F, Wang Z, Pokorny KS, Reinach PS. Differential dependence of regulatory volume decrease behavior in rabbit corneal epithelial cells on MAPK superfamily activation. Exp Eye Res 2007; 84:978-90. [PMID: 17397832 PMCID: PMC2747597 DOI: 10.1016/j.exer.2007.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2006] [Revised: 01/31/2007] [Accepted: 02/02/2007] [Indexed: 12/18/2022]
Abstract
We characterized the dependence of hypotonicity-induced regulatory volume decrease (RVD) responses on mitogen-activated protein kinase (MAPK) pathway signaling in SV40-immortalized rabbit corneal epithelial cells (RCEC). Following calcein-AM loading, RVD was monitored using a microplate fluorescence reader. Western blot analysis determined MAPK activation. After 30 min, the RVD response restored the relative cell volume to nearly isotonic values, whereas it was inhibited when cells were bathed either in a Cl- -free solution or with the Cl- -channel inhibitors: 5-nitro-2-(3-phenylpropylamino)benzoic acid or niflumic acid. Similar declines occurred with either a high-K+ (20 mM) supplemented solution or the K+ channel inhibitor 4-aminopyridine. Activation of extracellular signal-regulated kinase (ERK), p38, and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) was time and tonicity-dependent. Stimulation of ERK and SAPK/JNK was maximized earlier than that of p38. Activation of ERK and SAPK/JNK was insensitive to Cl- and K+ channel inhibitors, whereas inhibition with either PD98059 or SP600125, respectively, blocked RVD. However, inhibition of p38 with SB203580had no effect on RVD. Suppression of RVD instead blocked p38 activation. Differences in the dependence of RVD activation on Erk1/2 and p38 signaling were validated in dominant negative (d/n)-Erk1 and d/n-p38 cells. Volume-sensitive Cl- and K+ channel activation contributes, in concert, to RVD in RCEC. Therefore, swelling-induced ERK and SAPK/JNK stimulation precedes Cl- and K+ channel activation, whereas p38 activation occurs as a consequence of RVD.
Collapse
Affiliation(s)
- Zan Pan
- Department of Biological Sciences, State College of Optometry, State University of New York, New York, NY 10036, USA
| | - José E. Capó-Aponte
- Department of Biological Sciences, State College of Optometry, State University of New York, New York, NY 10036, USA
| | - Fan Zhang
- Department of Biological Sciences, State College of Optometry, State University of New York, New York, NY 10036, USA
| | - Zheng Wang
- Department of Biological Sciences, State College of Optometry, State University of New York, New York, NY 10036, USA
| | - Kathryn S. Pokorny
- The Institute of Ophthalmology and Visual Science, University of Medicine & Dentistry, New Jersey Medical School, Newark. NJ 07101
| | - Peter S. Reinach
- Department of Biological Sciences, State College of Optometry, State University of New York, New York, NY 10036, USA
| |
Collapse
|
91
|
Grondin M, Marion M, Denizeau F, Averill-Bates DA. Tributyltin induces apoptotic signaling in hepatocytes through pathways involving the endoplasmic reticulum and mitochondria. Toxicol Appl Pharmacol 2007; 222:57-68. [PMID: 17512566 DOI: 10.1016/j.taap.2007.03.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 03/15/2007] [Accepted: 03/26/2007] [Indexed: 10/23/2022]
Abstract
Tri-n-butyltin is a widespread environmental toxicant, which accumulates in the liver. This study investigates whether tri-n-butyltin induces pro-apoptotic signaling in rat liver hepatocytes through pathways involving the endoplasmic reticulum and mitochondria. Tri-n-butyltin activated the endoplasmic reticulum pathway of apoptosis, which was demonstrated by the activation of the protease calpain, its translocation to the plasma membrane, followed by cleavage of the calpain substrates, cytoskeletal protein vinculin, and caspase-12. Caspase-12 is localized to the cytoplasmic side of the endoplasmic reticulum and is involved in apoptosis mediated by the endoplasmic reticulum. Tri-n-butyltin also caused translocation of the pro-apoptotic proteins Bax and Bad from the cytosol to mitochondria, as well as changes in mitochondrial membrane permeability, events which can activate the mitochondrial death pathway. Tri-n-butyltin induced downstream apoptotic events in rat hepatocytes at the nuclear level, detected by chromatin condensation and by confocal microscopy using acridine orange. We investigated whether the tri-n-butyltin-induced pro-apoptotic events in hepatocytes could be linked to perturbation of intracellular calcium homeostasis, using confocal microscopy. Tri-n-butyltin caused changes in intracellular calcium distribution, which were similar to those induced by thapsigargin. Calcium was released from a subcellular compartment, which is likely to be the endoplasmic reticulum, into the cytosol. Cytosolic acidification, which is known to trigger apoptosis, also occurred and involved the Cl(-)/HCO(3)(-) exchanger. Pro-apoptotic events in hepatocytes were inhibited by the calcium chelator, Bapta-AM, and by a calpain inhibitor, which suggests that changes in intracellular calcium homeostasis are involved in tri-n-butyltin-induced apoptotic signaling in rat hepatocytes.
Collapse
Affiliation(s)
- Mélanie Grondin
- Département de Chimie, Centre de Recherche en Toxicologie de l'environnement (TOXEN), Université du Québec à Montréal, CP 8888, Succursale Centre-Ville, Montreal, Québec, Canada H3C 3P8
| | | | | | | |
Collapse
|
92
|
Sottosanto JB, Saranga Y, Blumwald E. Impact of AtNHX1, a vacuolar Na+/H+ antiporter, upon gene expression during short- and long-term salt stress in Arabidopsis thaliana. BMC PLANT BIOLOGY 2007; 7:18. [PMID: 17411438 PMCID: PMC1853094 DOI: 10.1186/1471-2229-7-18] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2006] [Accepted: 04/05/2007] [Indexed: 05/14/2023]
Abstract
BACKGROUND AtNHX1, the most abundant vacuolar Na+/H+ antiporter in Arabidopsis thaliana, mediates the transport of Na+ and K+ into the vacuole, influencing plant development and contributing to salt tolerance. In this report, microarray expression profiles of wild type plants, a T-DNA insertion knockout mutant of AtNHX1 (nhx1), and a 'rescued' line (NHX1::nhx1) were exposed to both short (12 h and 48 h) and long (one and two weeks) durations of a non-lethal salt stress to identify key gene transcripts associated with the salt response that are influenced by AtNHX1. RESULTS 147 transcripts showed both salt responsiveness and a significant influence of AtNHX1. Fifty-seven of these genes showed an influence of the antiporter across all salt treatments, while the remaining genes were influenced as a result of a particular duration of salt stress. Most (69%) of the genes were up-regulated in the absence of AtNHX1, with the exception of transcripts encoding proteins involved with metabolic and energy processes that were mostly down-regulated. CONCLUSION While part of the AtNHX1-influenced transcripts were unclassified, other transcripts with known or putative roles showed the importance of AtNHX1 to key cellular processes that were not necessarily limited to the salt stress response; namely calcium signaling, sulfur metabolism, cell structure and cell growth, as well as vesicular trafficking and protein processing. Only a small number of other salt-responsive membrane transporter transcripts appeared significantly influenced by AtNHX1.
Collapse
Affiliation(s)
- Jordan B Sottosanto
- Department of Plant Sciences, University of California, One Shields Ave, Davis, CA 95616, USA
| | - Yehoshua Saranga
- Department of Plant Sciences, University of California, One Shields Ave, Davis, CA 95616, USA
| | - Eduardo Blumwald
- Department of Plant Sciences, University of California, One Shields Ave, Davis, CA 95616, USA
| |
Collapse
|
93
|
Peluso I, Fina D, Caruso R, Stolfi C, Caprioli F, Fantini MC, Caspani G, Grossi E, Di Iorio L, Paone FM, Pallone F, Monteleone G. Lactobacillus paracasei subsp. paracasei B21060 suppresses human T-cell proliferation. Infect Immun 2007; 75:1730-1737. [PMID: 17242060 PMCID: PMC1865705 DOI: 10.1128/iai.01172-06] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 11/05/2006] [Accepted: 01/09/2007] [Indexed: 02/08/2023] Open
Abstract
Recent studies have shown that probiotics are beneficial in T-cell-mediated inflammatory diseases. The molecular mechanism by which probiotics work remains elusive, but accumulating evidence indicates that probiotics can modulate immune cell responses. Since T cells express receptors for bacterial products or components, we examined whether different strains of lactobacilli directly regulate the functions of human T cells. CD4(+) T cells were isolated from blood and intestinal lamina propria (LP) of normal individuals and patients with inflammatory bowel disease (IBD). Mononuclear cells were also isolated from Peyer's patches. Cells were activated with anti-CD3/CD2/CD28 in the presence or absence of Lactobacillus paracasei subsp. paracasei B21060, L. paracasei subsp. paracasei F19, or L. casei subsp. casei DG. Cell proliferation and death, Foxp3, intracellular pH, and cytokine production were evaluated by flow cytometry. We showed that L. paracasei subsp. paracasei B21060 but neither L. paracasei subsp. paracasei F19 nor L. casei subsp. casei DG inhibited blood CD4(+) T-cell growth. This effect was associated with no change in cell survival, expression of Foxp3, or production of gamma interferon, interleukin-4 (IL-4), IL-5, and IL-10. L. paracasei subsp. paracasei B21060-mediated blockade of CD4(+) T-cell proliferation required a viable bacterium and was associated with decreased MCT-1 expression and low intracellular pH. L. paracasei subsp. paracasei B21060 also inhibited the growth of Peyer's patch mononuclear cells, normal lymphocytes, and IBD CD4(+) LP lymphocytes without affecting cytokine production. The data show that L. paracasei subsp. paracasei B21060 blocks T-cell growth, thus suggesting a mechanism by which these probiotics could interfere with T-cell-driven immune responses.
Collapse
Affiliation(s)
- Ilaria Peluso
- Dipartimento di Medicina Interna, Università Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Bao Q, Lu W, Rabinowitz JD, Shi Y. Calcium blocks formation of apoptosome by preventing nucleotide exchange in Apaf-1. Mol Cell 2007; 25:181-92. [PMID: 17244527 DOI: 10.1016/j.molcel.2006.12.013] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2006] [Revised: 10/31/2006] [Accepted: 12/09/2006] [Indexed: 11/24/2022]
Abstract
Apaf-1 plays an essential role in apoptosis. In the presence of cytochrome c and dATP, Apaf-1 assembles into an oligomeric apoptosome, which is responsible for the activation of procaspase-9 and the maintenance of the enzymatic activity of the processed caspase-9. Regulation of apoptosome assembly by other cellular factors is poorly understood. Here we report that physiological concentrations of calcium ion negatively affect the assembly of apoptosome by inhibiting nucleotide exchange in the monomeric, autoinhibited Apaf-1 protein. Consequently, calcium blocks the ability of Apaf-1 to activate caspase-9. These observations suggest an important role of calcium homeostasis on the Apaf-1-dependent apoptotic pathway.
Collapse
Affiliation(s)
- Qing Bao
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ 08544, USA
| | | | | | | |
Collapse
|
95
|
Porichis F, Vlata Z, Hatzidakis G, Spandidos DA, Krambovitis E. HIV-1 gp120/V3-derived epitopes promote activation-induced cell death to superantigen-stimulated CD4+/CD45RO+ T cells. Immunol Lett 2007; 108:97-102. [PMID: 17141881 DOI: 10.1016/j.imlet.2006.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 11/03/2006] [Accepted: 11/05/2006] [Indexed: 01/07/2023]
Abstract
The third hypervirable (V3) domain of the HIV-1 envelope glycoprotein gp120 has been implicated in HIV pathogenesis via co-receptor usage of chemokine receptors CCR5 and CXCR4. As the protagonist cell populations in the asymptomatic phase of HIV-1 infection are infected macrophages and effector/memory (CD45RO+) CD4+ T cells that express CCR5, we established an in vitro model using human primary monocyte-derived macrophages and lymphocytes to investigate the role of V3 in affecting antigen presentation. We used staphylococcal enterotoxin A (SEA) as a superantigen at a low concentration of 1ng/ml, to activate naïve CD4+ T cells. Exposure of cells to SEA and lipoV3-liposomes increased the percentage of CD4+/CD45RO+/CCR5+ T cell population as compared to cells treated with SEA and plain liposomes. A consequent decrease of the percentage of CD4+/CD45RO+/CXCR4+ subset was observed. The V3-mediated activation was competitively inhibited by soluble V3-derived peptides with higher cationic charge. V3 enhanced also apoptosis as demonstrated by flow cytometry and intracellular calcium ion assays. These results reinforce the postulation that V3 alters the antigen presentation function itself, independent of specific antigens, thus leading to an enhanced activation-induced cell death (AICD) of responding T cells.
Collapse
Affiliation(s)
- Filippos Porichis
- Department of Applied Biochemistry and Immunology, Institute of Molecular Biology and Biotechnology, Vassilika Vouton, Heraklion, Crete, Greece
| | | | | | | | | |
Collapse
|
96
|
Waibel M, Kramer S, Lauber K, Lupescu A, Manns J, Schulze-Osthoff K, Lang F, Wesselborg S. Mitochondria are not required for death receptor-mediated cytosolic acidification during apoptosis. Apoptosis 2006; 12:623-30. [PMID: 17195091 DOI: 10.1007/s10495-006-0006-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Accepted: 11/01/2006] [Indexed: 11/29/2022]
Abstract
In addition to cell shrinkage, membrane blebbing, DNA fragmentation and phosphatidylserine exposure, intracellular acidification represents a hallmark of apoptosis. Although the mechanisms underlying cytosolic acidification during apoptosis remained largely elusive, a pivotal role of mitochondria has been proposed. In order to investigate the involvement of mitochondria in cytosolic acidification during apoptosis, we blocked the mitochondrial death pathway by overexpression of Bcl-2 and subsequently activated the death receptor pathway by anti-CD95 or TRAIL or the mitochondrial pathway by staurosporine. We show that Bcl-2 but not caspase inhibition prevented staurosporine-induced intracellular acidification. Thus, intracellular acidification in mitochondrial apoptosis is a Bcl-2-inhibitable, but caspase-independent process. In contrast, Bcl-2 only slightly delayed, but did not prevent intracellular acidification upon triggering of death receptors. The Na(+)/H(+) exchanger NHE1 was partially degraded during apoptosis but only to a small extent and and at a delayed time point when cytosolic acidification was almost completed. We therefore conclude that cytosolic acidification is mitochondrially controlled in response to mitochondria-dependent death stimuli, but requires additional caspase-dependent mechanisms during death receptor-mediated apoptosis.
Collapse
Affiliation(s)
- Michaela Waibel
- Department of Internal Medicine I, Eberhard-Karls-University, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Tani D, Monteilh-Zoller MK, Fleig A, Penner R. Cell cycle-dependent regulation of store-operated I(CRAC) and Mg2+-nucleotide-regulated MagNuM (TRPM7) currents. Cell Calcium 2006; 41:249-60. [PMID: 17064762 PMCID: PMC5663638 DOI: 10.1016/j.ceca.2006.07.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2006] [Revised: 07/07/2006] [Accepted: 07/07/2006] [Indexed: 12/30/2022]
Abstract
Calcium signaling is a central mechanism for numerous cellular functions and particularly relevant for immune cell proliferation. However, the role of calcium influx in mitotic cell cycle progression is largely unknown. We here report that proliferating rat mast cells RBL-2H3 tightly control their major store-operated calcium influx pathway, I(CRAC), during cell cycle progression. While I(CRAC) is maintained at control levels during the first gap phase (G1), the current is significantly up-regulated in preparation for and during chromatin duplication. However, mitosis strongly suppresses I(CRAC). Non-proliferating cells deprived of growth hormones strongly down-regulate I(CRAC) while increasing cell volume. We further show that the other known calcium (and magnesium) influx pathway in mast cells, the TRPM7-like magnesium-nucleotide-regulated metal (MagNuM) current, is largely uncoupled from cell cycle regulation except in G1. Taken together, our results demonstrate that both store-operated calcium influx via I(CRAC) and MagNuM are regulated at crucial checkpoints during cell cycle progression.
Collapse
Affiliation(s)
| | | | | | - Reinhold Penner
- Corresponding author. Tel.: +1 808 585 5366; fax: +1 808 585 5377. (R. Penner)
| |
Collapse
|
98
|
Timpe JM, Verrill KC, Black BN, Ding HF, Trempe JP. Adeno-associated virus induces apoptosis during coinfection with adenovirus. Virology 2006; 358:391-401. [PMID: 17011012 PMCID: PMC1839828 DOI: 10.1016/j.virol.2006.08.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2006] [Revised: 08/11/2006] [Accepted: 08/23/2006] [Indexed: 12/20/2022]
Abstract
Adeno-associated virus (AAV) is a nonpathogenic parvovirus that efficiently replicates in the presence of adenovirus (Ad). Exogenous expression of the AAV replication proteins induces caspase-dependent apoptosis, but determining if AAV infection causes apoptosis during viral infection is complicated by Ad-mediated programmed cell death. To eliminate Ad-induced cytolysis, we used an E3 adenoviral death protein (ADP) mutant, pm534. AAV and pm534-coinfected cells exhibited increased cell killing compared to pm534 alone. Relative to cells infected with Ad alone, AAV and wild-type Ad-infected cells displayed decreased ADP expression, increased cytolysis until the third day of the infection, and decreased cytolysis thereafter. Biochemical and morphological characteristics of apoptosis were observed during coinfections with AAV and pm534 or Ad, including a moderate degree of caspase activation that was not present during infections with pm534 or Ad alone. AAV coinfection also increased extracellular pH. These studies suggest that AAV induces caspase-dependent and caspase-independent apoptosis.
Collapse
Affiliation(s)
- Jennifer M Timpe
- Department of Biochemistry and Cancer Biology, Medical University of Ohio, 3035 Arlington Ave., Toledo, OH 43614-5804, USA
| | | | | | | | | |
Collapse
|
99
|
Klausen TK, Bergdahl A, Hougaard C, Christophersen P, Pedersen SF, Hoffmann EK. Cell cycle-dependent activity of the volume- and Ca2+-activated anion currents in Ehrlich lettre ascites cells. J Cell Physiol 2006; 210:831-42. [PMID: 17111356 DOI: 10.1002/jcp.20918] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recent evidence implicates the volume-regulated anion current (VRAC) and other anion currents in control or modulation of cell cycle progression; however, the precise involvement of anion channels in this process is unclear. Here, Cl- currents in Ehrlich Lettre Ascites (ELA) cells were monitored during cell cycle progression, under three conditions: (i) after osmotic swelling (i.e., VRAC), (ii) after an increase in the free intracellular Ca2+ concentration (i.e., the Ca2+-activated Cl- current, CaCC), and (iii) under steady-state isotonic conditions. The maximal swelling-activated VRAC current decreased in G1 and increased in early S phase, compared to that in G0. The isotonic steady-state current, which seems to be predominantly VRAC, also decreased in G1, and increased again in early S phase, to a level similar to that in G0. In contrast, the maximal CaCC current (500 nM free Ca2+ in the pipette), was unaltered from G0 to G1, but decreased in early S phase. A novel high-affinity anion channel inhibitor, the acidic di-aryl-urea NS3728, which inhibited both VRAC and CaCC, attenuated ELA cell growth, suggesting a possible mechanistic link between cell cycle progression and cell cycle-dependent changes in the capacity for conductive Cl- transport. It is suggested that in ELA cells, entrance into the S phase requires an increase in VRAC activity and/or an increased potential for regulatory volume decrease (RVD), and at the same time a decrease in CaCC magnitude.
Collapse
Affiliation(s)
- Thomas Kjaer Klausen
- Department of Biochemistry, Institute for Molecular Biology and Physiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|