Ionizing radiation, ion transports, and radioresistance of cancer cells

Stephan M Huber, Lena Butz, Benjamin Stegen, Dominik Klumpp, Norbert Braun, Peter Ruth, Franziska Eckert, Stephan M Huber, Lena Butz, Benjamin Stegen, Dominik Klumpp, Norbert Braun, Peter Ruth, Franziska Eckert

Abstract

The standard treatment of many tumor entities comprises fractionated radiation therapy which applies ionizing radiation to the tumor-bearing target volume. Ionizing radiation causes double-strand breaks in the DNA backbone that result in cell death if the number of DNA double-strand breaks exceeds the DNA repair capacity of the tumor cell. Ionizing radiation reportedly does not only act on the DNA in the nucleus but also on the plasma membrane. In particular, ionizing radiation-induced modifications of ion channels and transporters have been reported. Importantly, these altered transports seem to contribute to the survival of the irradiated tumor cells. The present review article summarizes our current knowledge on the underlying mechanisms and introduces strategies to radiosensitize tumor cells by targeting plasma membrane ion transports.

Keywords: DNA repair; cell cycle; ion channels; radiation therapy.

Figures

Figure 1
Figure 1
Cellular stress such as ionizing radiation induces the switch from “Grow” to “Go” phenotype.
Figure 2
Figure 2
Ionizing radiation (IR)-induced activation of BK K+ channels results in evasion of glioblastoma cells from radiation stress. Activation of BK channels occurs upstream of CaMKII and ClC-3 Cl− channels. The Na+/K+/2Cl− cotransporter accumulates Cl− above its electrochemical equilibrium. Cl− is used as osmolyte for volume decrease.
Figure 3
Figure 3
Mitochondrial ROS formation in dependence on ΔΨm (I, II, III, and IV, electron transport complexes I, II, III, and IV; Q: semiquinone radical; cyt C: cytochrome C). Ca2+ and the ATP/ADP ratio regulate the electron chain at complexes I and IV.

References

    1. Anderson E. J., Yamazaki H., Neufer P. D. (2007). Induction of endogenous uncoupling protein 3 suppresses mitochondrial oxidant emission during fatty acid-supported respiration. J. Biol. Chem. 282, 31257–31266 10.1074/jbc.M706129200
    1. Arcangeli A. (2011). Ion channels and transporters in cancer. 3. Ion channels in the tumor cell-microenvironment cross talk. Am. J. Physiol. Cell Physiol. 301, C762–C771
    1. Asuthkar S., Nalla A. K., Gondi C. S., Dinh D. H., Gujrati M., Mohanam S., et al. (2011). Gadd45a sensitizes medulloblastoma cells to irradiation and suppresses MMP-9-mediated EMT. Neuro Oncol. 13, 1059–1073 10.1093/neuonc/nor109
    1. Auperin A., Arriagada R., Pignon J. P., Le Pechoux C., Gregor A., Stephens R. J., et al. (1999). Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. prophylactic cranial irradiation overview collaborative group. N. Engl. J. Med. 341, 476–484 10.1056/NEJM199908123410703
    1. Ayyasamy V., Owens K. M., Desouki M. M., Liang P., Bakin A., Thangaraj K., et al. (2011). Cellular model of Warburg effect identifies tumor promoting function of UCP2 in breast cancer and its suppression by genipin. PLoS ONE 6:e24792 10.1371/journal.pone.0024792
    1. Badiga A. V., Chetty C., Kesanakurti D., Are D., Gujrati M., Klopfenstein J. D., et al. (2011). MMP-2 siRNA inhibits radiation-enhanced invasiveness in glioma cells. PLoS ONE 6:e20614 10.1371/journal.pone.0020614
    1. Baffy G. (2010). Uncoupling protein-2 and cancer. Mitochondrion 10, 243–252 10.1016/j.mito.2009.12.143
    1. Baffy G., Derdak Z., Robson S. C. (2011). Mitochondrial recoupling: a novel therapeutic strategy for cancer. Br. J. Cancer 105, 469–474
    1. Barendsen G. W. (1982). Dose fractionation, dose rate and iso-effect relationships for normal tissue responses. Int. J. Radiat. Oncol. Biol. Phys. 8, 1981–1997 10.1016/0360-301690459-X
    1. Bernier J., Hall E. J., Giaccia A. (2004). Radiation oncology: a century of achievements. Nat. Rev. Cancer 4, 737–747 10.1038/nrc1451
    1. Canazza A., Calatozzolo C., Fumagalli L., Bergantin A., Ghielmetti F., Fariselli L., et al. (2011). Increased migration of a human glioma cell line after in vitro CyberKnife irradiation. Cancer Biol. Ther. 12, 629–633 10.4161/cbt.12.7.16862
    1. Caraceni P., Ryu H. S., Van Thiel D. H., Borle A. B. (1995). Source of oxygen free radicals produced by rat hepatocytes during postanoxic reoxygenation. Biochim. Biophys. Acta 1268, 249–254 10.1016/0167-488900077-6
    1. Casneuf V. F., Fonteyne P., Van Damme N., Demetter P., Pauwels P., De Hemptinne B., et al. (2008). Expression of SGLT1, Bcl-2 and p53 in primary pancreatic cancer related to survival. Cancer Invest. 26, 852–859 10.1080/07357900801956363
    1. Catacuzzeno L., Aiello F., Fioretti B., Sforna L., Castigli E., Ruggieri P., et al. (2010). Serum-activated K and Cl currents underlay U87-MG glioblastoma cell migration. J. Cell Physiol. 226, 1926–1933 10.1002/jcp.22523
    1. Chakraborty M., Abrams S. I., Coleman C. N., Camphausen K., Schlom J., Hodge J. W. (2004). External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res. 64, 4328–4337 10.1158/0008-5472.CAN-04-0073
    1. Crompton M. (1999). The mitochondrial permeability transition pore and its role in cell death. Biochem. J. 341, 233–249
    1. Cuddapah V. A., Sontheimer H. (2010). Molecular interaction and functional regulation of ClC-3 by Ca2+/calmodulin-dependent protein kinase II (CaMKII) in human malignant glioma. J. Biol. Chem. 285, 11188–11196 10.1074/jbc.M109.097675
    1. Cullis P. M., Jones G. D. D., Lea J., Symons M. C. R., Sweeney M. (1987). The effects of ionizing radiation on deoxyribonucleic acid. Part 5. The role of thiols in chemical repair. J. Chem. Soc., Perkin Trans. 2, 1907–1914 10.1039/p29870001907
    1. Dale R. G. (1985). The application of the linear-quadratic dose-effect equation to fractionated and protracted radiotherapy. Br. J. Radiol. 58, 515–528 10.1259/0007-1285-58-690-515
    1. Dalla Pozza E., Fiorini C., Dando I., Menegazzi M., Sgarbossa A., Costanzo C., et al. (2012). Role of mitochondrial uncoupling protein 2 in cancer cell resistance to gemcitabine. Biochim. Biophys. Acta 1823, 1856–1863 10.1016/j.bbamcr.2012.06.007
    1. Dando I., Fiorini C., Pozza E. D., Padroni C., Costanzo C., Palmieri M., et al. (2013). UCP2 inhibition triggers ROS-dependent nuclear translocation of GAPDH and autophagic cell death in pancreatic adenocarcinoma cells. Biochim. Biophys. Acta 1833, 672–679 10.1016/j.bbamcr.2012.10.028
    1. Darby S., Mcgale P., Correa C., Taylor C., Arriagada R., Clarke M., et al. (2011). Effect of radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast cancer death: meta-analysis of individual patient data for 10, 801 women in 17 randomised trials. Lancet 378, 1707–1716 10.1016/S0140-673661629-2
    1. Demaria S., Formenti S. C. (2012). Radiation as an immunological adjuvant: current evidence on dose and fractionation. Front. Oncol. 2:153 10.3389/fonc.2012.00153
    1. Deng S., Yang Y., Han Y., Li X., Wang X., Li X., et al. (2012). UCP2 inhibits ROS-mediated apoptosis in A549 under hypoxic conditions. PLoS ONE 7:e30714 10.1371/journal.pone.0030714
    1. Derdak Z., Mark N. M., Beldi G., Robson S. C., Wands J. R., Baffy G. (2008). The mitochondrial uncoupling protein-2 promotes chemoresistance in cancer cells. Cancer Res. 68, 2813–2819 10.1158/0008-5472.CAN-08-0053
    1. Dittmann K., Mayer C., Kehlbach R., Rothmund M. C., Peter Rodemann H. (2009). Radiation-induced lipid peroxidation activates src kinase and triggers nuclear EGFR transport. Radiother. Oncol. 92, 379–382 10.1016/j.radonc.2009.06.003
    1. Dittmann K., Mayer C., Rodemann H. P., Huber S. M. (2013). EGFR cooperates with glucose transporter SGLT1 to enable chromatin remodeling in response to ionizing radiation. Radiother. Oncol. 107, 247–251 10.1016/j.radonc.2013.03.016
    1. Dorn G. W., 2nd. (2013). Molecular mechanisms that differentiate apoptosis from programmed necrosis. Toxicol. Pathol. 41, 227–234 10.1177/0192623312466961
    1. Echtay K. S., Roussel D., St-Pierre J., Jekabsons M. B., Cadenas S., Stuart J. A., et al. (2002). Superoxide activates mitochondrial uncoupling proteins. Nature 415, 96–99 10.1038/415096a
    1. Eckert F., Alloussi S., Paulsen F., Bamberg M., Zips D., Spillner P., et al. (2013). Prospective evaluation of a hydrogel spacer for rectal separation in dose-escalated intensity-modulated radiotherapy for clinically localized prostate cancer. BMC Cancer 13:27 10.1186/1471-2407-13-27
    1. Eckert F., Fehm T., Bamberg M., Muller A. C. (2010a). Small cell carcinoma of vulva: curative multimodal treatment in face of resistance to initial standard chemotherapy. Strahlenther. Onkol. 186, 521–524
    1. Eckert F., Matuschek C., Mueller A. C., Weinmann M., Hartmann J. T., Belka C., et al. (2010b). Definitive radiotherapy and single-agent radiosensitizing ifosfamide in patients with localized, irresectable soft tissue sarcoma: a retrospective analysis. Radiat. Oncol. 5, 55
    1. Eckert F., Gani C., Bamberg M., Muller A. C. (2012). Cerebral metastases in extrapulmonary cell carcinoma: implications for the use of prophylactic cranial irradiation. Strahlenther. Onkol. 188, 478–483 10.1007/s00066-012-0084-5
    1. Ernest N. J., Weaver A. K., Van Duyn L. B., Sontheimer H. W. (2005). Relative contribution of chloride channels and transporters to regulatory volume decrease in human glioma cells. Am. J. Physiol. Cell Physiol. 288, C1451–1460 10.1152/ajpcell.00503.2004
    1. Fink B. D., Hong Y. S., Mathahs M. M., Scholz T. D., Dillon J. S., Sivitz W. I. (2002). UCP2-dependent proton leak in isolated mammalian mitochondria. J. Biol. Chem. 277, 3918–3925 10.1074/jbc.M107955200
    1. Finkelstein S. E., Iclozan C., Bui M. M., Cotter M. J., Ramakrishnan R., Ahmed J., et al. (2012). Combination of external beam radiotherapy (EBRT) with intratumoral injection of dendritic cells as neo-adjuvant treatment of high-risk soft tissue sarcoma patients. Int. J. Radiat. Oncol. Biol. Phys. 82, 924–932 10.1016/j.ijrobp.2010.12.068
    1. Formenti S. C., Demaria S. (2013). Combining radiotherapy and cancer immunotherapy: a paradigm shift. J. Natl. Cancer Inst. 105, 256–265 10.1093/jnci/djs629
    1. Fuessel S., Meye A., Schmitz M., Zastrow S., Linne C., Richter K., et al. (2006). Vaccination of hormone-refractory prostate cancer patients with peptide cocktail-loaded dendritic cells: results of a phase I clinical trial. Prostate 66, 811–821 10.1002/pros.20404
    1. Fyles A., Milosevic M., Hedley D., Pintilie M., Levin W., Manchul L., et al. (2002). Tumor hypoxia has independent predictor impact only in patients with node-negative cervix cancer. J. Clin. Oncol. 20, 680–687 10.1200/JCO.20.3.680
    1. Fyles A., Milosevic M., Pintilie M., Syed A., Levin W., Manchul L., et al. (2006). Long-term performance of interstial fluid pressure and hypoxia as prognostic factors in cervix cancer. Radiother. Oncol. 80, 132–137 10.1016/j.radonc.2006.07.014
    1. Ganapathy V., Thangaraju M., Prasad P. D. (2009). Nutrient transporters in cancer: relevance to Warburg hypothesis and beyond. Pharmacol. Ther. 121, 29–40 10.1016/j.pharmthera.2008.09.005
    1. Garcia-Martinez C., Sibille B., Solanes G., Darimont C., Mace K., Villarroya F., et al. (2001). Overexpression of UCP3 in cultured human muscle lowers mitochondrial membrane potential, raises ATP/ADP ratio, and favors fatty acid vs. glucose oxidation. FASEB J. 15, 2033–2035
    1. German-Cancer-Aid. (2013a). Homepage. Available online at:
    1. German-Cancer-Aid. (2013b). Information Booklet. Available online at:
    1. Glenny A. M., Furness S., Worthington H. V., Conway D. I., Oliver R., Clarkson J. E., et al. (2010). Interventions for the treatment of oral cavity and oropharyngeal cancer: radiotherapy. Cochrane Database Syst. Rev. CD006387 10.1002/14651858.CD006387.pub2
    1. Gomez-Ospina N., Tsuruta F., Barreto-Chang O., Hu L., Dolmetsch R. (2006). The C terminus of the L-type voltage-gated calcium channel Ca(V)1.2 encodes a transcription factor. Cell 127, 591–606 10.1016/j.cell.2006.10.017
    1. Gonidi M., Athanassiadou A. M., Patsouris E., Tsipis A., Dimopoulos S., Kyriakidou V., et al. (2011). Mitochondrial UCP4 and bcl-2 expression in imprints of breast carcinomas: relationship with DNA ploidy and classical prognostic factors. Pathol. Res. Pract. 207, 377–382 10.1016/j.prp.2011.03.007
    1. Grills I. S., Hope A. J., Guckenberger M., Kestin L. L., Werner-Wasik M., Yan D., et al. (2012). A collaborative analysis of stereotactic lung radiotherapy outcomes for early-stage non-small-cell lung cancer using daily online cone-beam computed tomography image-guided radiotherapy. J. Thorac. Oncol. 7, 1382–1393 10.1097/JTO.0b013e318260e00d
    1. Gross G. J., Peart J. N. (2003). KATP channels and myocardial preconditioning: an update. Am. J. Physiol. Heart Circ. Physiol. 285, H921–H930
    1. Grosso J. F., Jure-Kunkel M. N. (2013). CTLA-4 blockade in tumor models: an overview of preclinical and translational research. Cancer Immun. 13, 5
    1. Haas B. R., Cuddapah V. A., Watkins S., Rohn K. J., Dy T. E., Sontheimer H. (2011). With-No-Lysine Kinase 3 (WNK3) stimulates glioma invasion by regulating cell volume. Am. J. Physiol. Cell Physiol. 301, C1150–C1160 10.1152/ajpcell.00203.2011
    1. Haas B. R., Sontheimer H. (2010). Inhibition of the sodium-potassium-chloride cotransporter isoform-1 reduces glioma invasion. Cancer Res. 70, 5597–5606 10.1158/0008-5472.CAN-09-4666
    1. Hanahan D., Weinberg R. A. (2011). Hallmarks of cancer: the next generation. Cell 144, 646–674 10.1016/j.cell.2011.02.013
    1. Harada H. (2011). How can we overcome tumor hypoxia in radiation therapy. J. Radiat. Res. 52, 545–556
    1. Harper M. E., Antoniou A., Villalobos-Menuey E., Russo A., Trauger R., Vendemelio M., et al. (2002). Characterization of a novel metabolic strategy used by drug-resistant tumor cells. FASEB J. 16, 1550–1557 10.1096/fj.02-0541com
    1. Hartung F., Stuhmer W., Pardo L. A. (2011). Tumor cell-selective apoptosis induction through targeting of K(V)10.1 via bifunctional TRAIL antibody. Mol. Cancer 10, 109
    1. Hatzikirou H., Basanta D., Simon M., Schaller K., Deutsch A. (2012). ‘Go or Grow’: the key to the emergence of invasion in tumour progression. Math. Med. Biol. 29, 49–65 10.1093/imammb/dqq011
    1. Heise N., Palme D., Misovic M., Koka S., Rudner J., Lang F., et al. (2010). Non-selective cation channel-mediated Ca2+-entry and activation of Ca2+/calmodulin-dependent kinase II contribute to G2/M cell cycle arrest and survival of irradiated leukemia cells. Cell Physiol. Biochem. 26, 597–608 10.1159/000322327
    1. Helmke B. M., Reisser C., Idzko M., Dyckhoff G., Herold-Mende C. (2004). Expression of SGLT-1 in preneoplastic and neoplastic lesions of the head and neck. Oral Oncol. 40, 28–35 10.1016/S1368-837500129-5
    1. Hockaday D. C., Shen S., Fiveash J., Raubitschek A., Colcher D., Liu A., et al. (2005). Imaging glioma extent with 131I-TM-601. J. Nucl. Med. 46, 580–586
    1. Horimoto M., Resnick M. B., Konkin T. A., Routhier J., Wands J. R., Baffy G. (2004). Expression of uncoupling protein-2 in human colon cancer. Clin. Cancer Res. 10, 6203–6207 10.1158/1078-0432.CCR-04-0419
    1. Horiot J. C., Le Fur R., N'guyen T., Chenal C., Schraub S., Alfonsi S., et al. (1992). Hyperfractionation versus conventional fractionation in oropharyngeal carcinoma: final analysis of a randomized trial of the EORTC cooperative group of radiotherapy. Radiother. Oncol. 25, 231–241 10.1016/0167-814090242-M
    1. Huber S. M. (2013). Oncochannels. Cell Calcium. 53, 241–255 10.1016/j.ceca.2013.01.001
    1. Huber S. M., Misovic M., Mayer C., Rodemann H. P., Dittmann K. (2012). EGFR-mediated stimulation of sodium/glucose cotransport promotes survival of irradiated human A549 lung adenocarcinoma cells. Radiother. Oncol. 103, 373–379 10.1016/j.radonc.2012.03.008
    1. Ishikawa N., Oguri T., Isobe T., Fujitaka K., Kohno N. (2001). SGLT gene expression in primary lung cancers and their metastatic lesions. Jpn. J. Cancer Res. 92, 874–879 10.1111/j.1349-7006.2001.tb01175.x
    1. Johnson J., Nowicki M. O., Lee C. H., Chiocca E. A., Viapiano M. S., Lawler S. E., et al. (2009). Quantitative analysis of complex glioma cell migration on electrospun polycaprolactone using time-lapse microscopy. Tissue Eng. Part C Methods 15, 531–540 10.1089/ten.tec.2008.0486
    1. Jones B., Dale R. G., Gaya A. M. (2006). Linear quadratic modeling of increased late normal-tissue effects in special clinical situations. Int. J. Radiat. Oncol. Biol. Phys. 64, 948–953 10.1016/j.ijrobp.2005.10.016
    1. Jung J. W., Hwang S. Y., Hwang J. S., Oh E. S., Park S., Han I. O. (2007). Ionising radiation induces changes associated with epithelial-mesenchymal transdifferentiation and increased cell motility of A549 lung epithelial cells. Eur. J. Cancer 43, 1214–1224 10.1016/j.ejca.2007.01.034
    1. Kadenbach B. (2003). Intrinsic and extrinsic uncoupling of oxidative phosphorylation. Biochim. Biophys. Acta 1604, 77–94 10.1016/S0005-272800027-6
    1. Kargiotis O., Chetty C., Gogineni V., Gondi C. S., Pulukuri S. M., Kyritsis A. P., et al. (2008). uPA/uPAR downregulation inhibits radiation-induced migration, invasion and angiogenesis in IOMM-Lee meningioma cells and decreases tumor growth in vivo. Int. J. Oncol. 33, 937–947
    1. Kil W. J., Tofilon P. J., Camphausen K. (2012). Post-radiation increase in VEGF enhances glioma cell motility in vitro. Radiat. Oncol. 7, 25 10.1186/1748-717X-7-25
    1. Kim J. S., Chang J. W., Yun H. S., Yang K. M., Hong E. H., Kim D. H., et al. (2010). Chloride intracellular channel 1 identified using proteomic analysis plays an important role in the radiosensitivity of HEp-2 cells via reactive oxygen species production. Proteomics 10, 2589–2604 10.1002/pmic.200900523
    1. Klotz L., Venier N., Colquhoun A. J., Sasaki H., Loblaw D. A., Fleshner N., et al. (2011). Capsaicin, a novel radiosensitizer, acts via a TRPV6 mediated phenomenon. J. Clin. Oncol. 29(Suppl. 7; Abstr. 23).
    1. Koebel C. M., Vermi W., Swann J. B., Zerafa N., Rodig S. J., Old L. J., et al. (2007). Adaptive immunity maintains occult cancer in an equilibrium state. Nature 450, 903–907 10.1038/nature06309
    1. Korshunov S. S., Skulachev V. P., Starkov A. A. (1997). High protonic potential actuates a mechanism of production of reactive oxygen species in mitochondria. FEBS Lett. 416, 15–18 10.1016/S0014-579301159-9
    1. Kotecha R., Yamada Y., Pei X., Kollmeier M. A., Cox B., Cohen G. N., et al. (2013). Clinical outcomes of high-dose-rate brachytherapy and external beam radiotherapy in the management of clinically localized prostate cancer. Brachytherapy 12, 44–49 10.1016/j.brachy.2012.05.003
    1. Kuai X. Y., Ji Z. Y., Zhang H. J. (2010). Mitochondrial uncoupling protein 2 expression in colon cancer and its clinical significance. World J. Gastroenterol. 16, 5773–5778 10.3748/wjg.v16.i45.5773
    1. Kuo S. S., Saad A. H., Koong A. C., Hahn G. M., Giaccia A. J. (1993). Potassium-channel activation in response to low doses of gamma-irradiation involves reactive oxygen intermediates in nonexcitatory cells. Proc. Natl. Acad. Sci. U.S.A. 90, 908–912 10.1073/pnas.90.3.908
    1. Langenbacher M., Abdel-Jalil R. J., Voelter W., Weinmann M., Huber S. M. (2013). In vitro hypoxic cytotoxicity and hypoxic radiosensitization. Efficacy of the novel 2-nitroimidazole N, N, N-tris[2-(2-nitro-1H-imidazol-1-yl)ethyl]amine. Strahlenther. Onkol. 189, 246–254 10.1007/s00066-012-0273-2
    1. Le Pechoux C., Laplanche A., Faivre-Finn C., Ciuleanu T., Wanders R., Lerouge D., et al. (2011). Clinical neurological outcome and quality of life among patients with limited small-cell cancer treated with two different doses of prophylactic cranial irradiation in the intergroup phase III trial (PCI99-01, EORTC 22003-08004, RTOG (0212). and IFCT 99-01). Ann. Oncol. 22, 1154–1163 10.1093/annonc/mdq576
    1. Leiprecht N., Munoz C., Alesutan I., Siraskar G., Sopjani M., Foller M., et al. (2011). Regulation of Na(+)-coupled glucose carrier SGLT1 by human papillomavirus 18 E6 protein. Biochem. Biophys. Res. Commun. 404, 695–700 10.1016/j.bbrc.2010.12.044
    1. Li C., Jackson R. M. (2002). Reactive species mechanisms of cellular hypoxia-reoxygenation injury. Am. J. Physiol. Cell Physiol. 282, C227–241 10.1152/ajpcell.00112.2001
    1. Liu X., Chang Y., Reinhart P. H., Sontheimer H., Chang Y. (2002). Cloning and characterization of glioma BK, a novel BK channel isoform highly expressed in human glioma cells. J. Neurosci. 22, 1840–1849
    1. Lobikin M., Chernet B., Lobo D., Levin M. (2012). Resting potential, oncogene-induced tumorigenesis, and metastasis: the bioelectric basis of cancer in vivo. Phys. Biol. 9, 065002 10.1088/1478-3975/9/6/065002
    1. Lu Z., Sack M. N. (2008). ATF-1 is a hypoxia-responsive transcriptional activator of skeletal muscle mitochondrial-uncoupling protein 3. J. Biol. Chem. 283, 23410–23418 10.1074/jbc.M801236200
    1. Lui V. C., Lung S. S., Pu J. K., Hung K. N., Leung G. K. (2010). Invasion of human glioma cells is regulated by multiple chloride channels including ClC-3. Anticancer Res. 30, 4515–4524
    1. Lutz S. (2007). Palliative whole-brain radiotherapy fractionation: convenience versus cognition. Cancer 110, 2363–2365
    1. Lyons S. A., O'neal J., Sontheimer H. (2002). Chlorotoxin, a scorpion-derived peptide, specifically binds to gliomas and tumors of neuroectodermal origin. Glia 39, 162–173 10.1002/glia.10083
    1. Maftei C. A., Bayer C., Shi K., Astner S. T., Vaupel P. (2011). Changes in the fraction of total hypoxia and hypoxia subtypes in human squamous cell carcinomas upon fractionated irradiation: evaluation using pattern recognition in microcirculatory supply units. Radiother. Oncol. 101, 209–216 10.1016/j.radonc.2011.05.023
    1. Mailloux R. J., Adjeitey C. N., Harper M. E. (2010). Genipin-induced inhibition of uncoupling protein-2 sensitizes drug-resistant cancer cells to cytotoxic agents. PLoS ONE 5:e13289 10.1371/journal.pone.0013289
    1. Mamelak A. N., Jacoby D. B. (2007). Targeted delivery of antitumoral therapy to glioma and other malignancies with synthetic chlorotoxin (TM-601). Expert Opin. Drug Deliv. 4, 175–186 10.1517/17425247.4.2.175
    1. Marks L. B., Dewhirst M. (1991). Accelerated repopulation: friend or foe. Exploiting changes in tumor growth characteristics to improve the “efficiency” of radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 21, 1377–1383 10.1016/0360-301690301-J
    1. Masumoto K., Tsukimoto M., Kojima S. (2013). Role of TRPM2 and TRPV1 cation channels in cellular responses to radiation-induced DNA damage. Biochim. Biophys. Acta 1830, 3382–3390 10.1016/j.bbagen.2013.02.020
    1. Mccoy E. S., Haas B. R., Sontheimer H. (2010). Water permeability through aquaporin-4 is regulated by protein kinase C and becomes rate-limiting for glioma invasion. Neuroscience 168, 971–981 10.1016/j.neuroscience.2009.09.020
    1. Mccoy E., Sontheimer H. (2007). Expression and function of water channels (aquaporins) in migrating malignant astrocytes. Glia 55, 1034–1043 10.1002/glia.20524
    1. Mcferrin M. B., Sontheimer H. (2006). A role for ion channels in glioma cell invasion. Neuron Glia Biol. 2, 39–49 10.1017/S1740925X06000044
    1. Mcleod C. J., Aziz A., Hoyt R. F., Jr., Mccoy J. P., Jr., Sack M. N. (2005). Uncoupling proteins 2 and 3 function in concert to augment tolerance to cardiac ischemia. J. Biol. Chem. 280, 33470–33476 10.1074/jbc.M505258200
    1. Montana V., Sontheimer H. (2011). Bradykinin promotes the chemotactic invasion of primary brain tumors. J. Neurosci. 31, 4858–4867 10.1523/JNEUROSCI.3825-10.2011
    1. Muller A. C., Eckert F., Heinrich V., Bamberg M., Brucker S., Hehr T. (2011). Re-surgery and chest wall re-irradiation for recurrent breast cancer: a second curative approach. BMC Cancer 11:197 10.1186/1471-2407-11-197
    1. Muller A. C., Gani C., Weinmann M., Mayer F., Sipos B., Bamberg M., et al. (2012). Limited disease of extra-pulmonary small cell carcinoma. Impact of local treatment and nodal status, role of cranial irradiation. Strahlenther. Onkol. 188, 269–273 10.1007/s00066-011-0045-4
    1. Murata M., Akao M., O'rourke B., Marban E. (2001). Mitochondrial ATP-sensitive potassium channels attenuate matrix Ca(2+) overload during simulated ischemia and reperfusion: possible mechanism of cardioprotection. Circ. Res. 89, 891–898 10.1161/hh2201.100205
    1. Nalla A. K., Asuthkar S., Bhoopathi P., Gujrati M., Dinh D. H., Rao J. S. (2010). Suppression of uPAR retards radiation-induced invasion and migration mediated by integrin beta1/FAK signaling in medulloblastoma. PLoS ONE 5:e13006 10.1371/journal.pone.0013006
    1. Narita T., Aoyama H., Hirata K., Onodera S., Shiga T., Kobayashi H., et al. (2012). Reoxygenation of glioblastoma multiforme treated with fractionated radiotherapy concomitant with temozolomide: changes defined by 18F-fluoromisonidazole positron emission tomography: two case reports. Jpn. J. Clin. Oncol. 42, 120–123 10.1093/jjco/hyr181
    1. Nelson J. A., Falk R. E. (1993). The efficacy of phloridzin and phloretin on tumor cell growth. Anticancer Res. 13, 2287–2292
    1. Nguyen L. N., Ang K. K. (2002). Radiotherapy for cancer of the head and neck: altered fractionation regimens. Lancet Oncol. 3, 693–701 10.1016/S1470-204500906-3
    1. Niyazi M., Siefert A., Schwarz S. B., Ganswindt U., Kreth F. W., Tonn J. C., et al. (2011). Therapeutic options for recurrent malignant glioma. Radiother. Oncol. 98, 1–14 10.1016/j.radonc.2010.11.006
    1. Ohshima Y., Tsukimoto M., Harada H., Kojima S. (2012). Involvement of connexin43 hemichannel in ATP release after gamma-irradiation. J. Radiat. Res. 53, 551–557 10.1093/jrr/rrs014
    1. Oleinika K., Nibbs R. J., Graham G. J., Fraser A. R. (2013). Suppression, subversion and escape: the role of regulatory T cells in cancer progression. Clin. Exp. Immunol. 171, 36–45 10.1111/j.1365-2249.2012.04657.x
    1. Olsen M. L., Schade S., Lyons S. A., Amaral M. D., Sontheimer H. (2003). Expression of voltage-gated chloride channels in human glioma cells. J. Neurosci. 23, 5572–5582
    1. Ozcan C., Palmeri M., Horvath T. L., Russell K. S., Russell R. R. (2013). Role of uncoupling protein 3 in ischemia-reperfusion injury, arrhythmias and preconditioning. Am. J. Physiol. Heart Circ. Physiol. 304, H1192–H1200 10.1152/ajpheart.00592.2012
    1. Pajonk F., Vlashi E., Mcbride W. H. (2010). Radiation resistance of cancer stem cells: the 4 R's of radiobiology revisited. Stem Cells 28, 639–648 10.1002/stem.318
    1. Palme D., Misovic M., Schmid E., Klumpp D., Salih H. R., Rudner J., et al. (2013). Kv3.4 potassium channel-mediated electrosignaling controls cell cycle and survival of irradiated leukemia cells. Pflugers Archiv 465, 1209–1221 10.1007/s00424-013-1249-5
    1. Pawlik T. M., Keyomarsi K. (2004). Role of cell cycle in mediating sensitivity to radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 59, 928–942 10.1016/j.ijrobp.2004.03.005
    1. Pfefferle A., Mailloux R. J., Adjeitey C. N., Harper M. E. (2012). Glutathionylation of UCP2 sensitizes drug resistant leukemia cells to chemotherapeutics. Biochim. Biophys. Acta 1833, 80–89 10.1016/j.bbamcr.2012.10.006
    1. Pickhard A. C., Margraf J., Knopf A., Stark T., Piontek G., Beck C., et al. (2011). Inhibition of radiation induced migration of human head and neck squamous cell carcinoma cells by blocking of EGF receptor pathways. BMC Cancer 11:388 10.1186/1471-2407-11-388
    1. Pizzo P., Drago I., Filadi R., Pozzan T. (2012). Mitochondrial Ca(2)(+) homeostasis: mechanism, role, and tissue specificities. Pflugers Arch. 464, 3–17 10.1007/s00424-012-1122-y
    1. Pons D. G., Sastre-Serra J., Nadal-Serrano M., Oliver A., Garcia-Bonafe M., Bover I., et al. (2012). Initial activation status of the antioxidant response determines sensitivity to carboplatin/paclitaxel treatment of ovarian cancer. Anticancer Res. 32, 4723–4728
    1. Prasad A., Stone G. W., Holmes D. R., Gersh B. (2009). Reperfusion injury, microvascular dysfunction, and cardioprotection: the “dark side” of reperfusion. Circulation 120, 2105–2112 10.1161/CIRCULATIONAHA.108.814640
    1. Rades D., Kieckebusch S., Lohynska R., Veninga T., Stalpers L. J., Dunst J., et al. (2007a). Reduction of overall treatment time in patients irradiated for more than three brain metastases. Int. J. Radiat. Oncol. Biol. Phys. 69, 1509–1513
    1. Rades D., Lohynska R., Veninga T., Stalpers L. J., Schild S. E. (2007b). Evaluation of 2 whole-brain radiotherapy schedules and prognostic factors for brain metastases in breast cancer patients. Cancer 110, 2587–2592
    1. Ransom C. B., Liu X., Sontheimer H. (2002). BK channels in human glioma cells have enhanced calcium sensitivity. Glia 38, 281–291 10.1002/glia.10064
    1. Ransom C. B., Sontheimer H. (2001). BK channels in human glioma cells. J. Neurophysiol. 85, 790–803
    1. Rasola A., Bernardi P. (2011). Mitochondrial permeability transition in Ca(2+)-dependent apoptosis and necrosis. Cell Calcium 50, 222–233 10.1016/j.ceca.2011.04.007
    1. Rieken S., Habermehl D., Mohr A., Wuerth L., Lindel K., Weber K., et al. (2011). Targeting alphanubeta3 and alphanubeta5 inhibits photon-induced hypermigration of malignant glioma cells. Radiat. Oncol. 6, 132 10.1186/1748-717X-6-132
    1. Robbins D., Zhao Y. (2011). New aspects of mitochondrial uncoupling proteins (UCPs) and their roles in tumorigenesis. Int. J. Mol. Sci. 12, 5285–5293 10.3390/ijms12085285
    1. Rodrigues G., Zindler J., Warner A., Lagerwaard F. (2013). Recursive partitioning analysis for the prediction of stereotactic radiosurgery brain metastases lesion control. Oncologist 18, 330–335 10.1634/theoncologist.2012-0316
    1. Ruggieri P., Mangino G., Fioretti B., Catacuzzeno L., Puca R., Ponti D., et al. (2012). The inhibition of KCa3.1 channels activity reduces cell motility in glioblastoma derived cancer stem cells. PLoS ONE 7:e47825 10.1371/journal.pone.0047825
    1. Sack M. N. (2006). Mitochondrial depolarization and the role of uncoupling proteins in ischemia tolerance. Cardiovasc. Res. 72, 210–219 10.1016/j.cardiores.2006.07.010
    1. Samudio I., Fiegl M., Mcqueen T., Clise-Dwyer K., Andreeff M. (2008). The warburg effect in leukemia-stroma cocultures is mediated by mitochondrial uncoupling associated with uncoupling protein 2 activation. Cancer Res. 68, 5198–5205 10.1158/0008-5472.CAN-08-0555
    1. Santandreu F. M., Valle A., Fernandez De Mattos S., Roca P., Oliver J. (2009). Hydrogen peroxide regulates the mitochondrial content of uncoupling protein 5 in colon cancer cells. Cell Physiol. Biochem. 24, 379–390 10.1159/000257430
    1. Sastre-Serra J., Nadal-Serrano M., Pons D. G., Valle A., Garau I., Garcia-Bonafe M., et al. (2013). The oxidative stress in breast tumors of postmenopausal women is ERalpha/ERbeta ratio dependent. Free Radic. Biol. Med. 61C, 11–17 10.1016/j.freeradbiomed.2013.03.005
    1. Sauer R., Liersch T., Merkel S., Fietkau R., Hohenberger W., Hess C., et al. (2012). Preoperative versus postoperative chemoradiotherapy for locally advanced rectal cancer: results of the German CAO/ARO/AIO-94 randomized phase III trial after a median follow-up of 11 years. J. Clin. Oncol. 30, 1926–1933 10.1200/JCO.2011.40.1836
    1. Schwab A., Fabian A., Hanley P. J., Stock C. (2012). Role of ion channels and transporters in cell migration. Physiol. Rev. 92, 1865–1913 10.1152/physrev.00018.2011
    1. Schwab A., Nechyporuk-Zloy V., Fabian A., Stock C. (2007). Cells move when ions and water flow. Pflugers Arch. 453, 421–432 10.1007/s00424-006-0138-6
    1. Sciaccaluga M., Fioretti B., Catacuzzeno L., Pagani F., Bertollini C., Rosito M., et al. (2010). CXCL12-induced glioblastoma cell migration requires intermediate conductance Ca2+-activated K+ channel activity. Am. J. Physiol. Cell Physiol. 299, C175–C184 10.1152/ajpcell.00344.2009
    1. Shields J. D., Kourtis I. C., Tomei A. A., Roberts J. M., Swartz M. A. (2010). Induction of lymphoidlike stroma and immune escape by tumors that express the chemokine CCL21. Science 328, 749–752 10.1126/science.1185837
    1. Singh H., Stefani E., Toro L. (2012). Intracellular BK(Ca) (iBK(Ca)) channels. J. Physiol. 590, 5937–5947 10.1113/jphysiol.2011.215533
    1. Skulachev V. P. (1998). Uncoupling: new approaches to an old problem of bioenergetics. Biochim. Biophys. Acta 1363, 100–124 10.1016/S0005-272800091-1
    1. Sofia Vala I., Martins L. R., Imaizumi N., Nunes R. J., Rino J., Kuonen F., et al. (2010). Low doses of ionizing radiation promote tumor growth and metastasis by enhancing angiogenesis. PLoS ONE 5:e11222 10.1371/journal.pone.0011222
    1. Sontheimer H. (2008). An unexpected role for ion channels in brain tumor metastasis. Exp. Biol. Med. (Maywood) 233, 779–791 10.3181/0711-MR-308
    1. Sreekumar A., Nyati M. K., Varambally S., Barrette T. R., Ghosh D., Lawrence T. S., et al. (2001). Profiling of cancer cells using protein microarrays: discovery of novel radiation-regulated proteins. Cancer Res. 61, 7585–7593
    1. Steinle M., Palme D., Misovic M., Rudner J., Dittmann K., Lukowski R., et al. (2011). Ionizing radiation induces migration of glioblastoma cells by activating BK K(+) channels. Radiother. Oncol. 101, 122–126 10.1016/j.radonc.2011.05.069
    1. Stock C., Schwab A. (2009). Protons make tumor cells move like clockwork. Pflugers Arch. 458, 981–992 10.1007/s00424-009-0677-8
    1. Stupp R., Van Den Bent M. J., Hegi M. E. (2005). Optimal role of temozolomide in the treatment of malignant gliomas. Curr. Neurol. Neurosci. Rep. 5, 198–206 10.1007/s11910-005-0047-7
    1. Su W. P., Lo Y. C., Yan J. J., Liao I. C., Tsai P. J., Wang H. C., et al. (2012). Mitochondrial uncoupling protein 2 regulates the effects of paclitaxel on Stat3 activation and cellular survival in lung cancer cells. Carcinogenesis 33, 2065–2075 10.1093/carcin/bgs253
    1. Szabo I., Leanza L., Gulbins E., Zoratti M. (2012). Physiology of potassium channels in the inner membrane of mitochondria. Pflugers Arch. 463, 231–246 10.1007/s00424-011-1058-7
    1. Talbot D. A., Brand M. D. (2005). Uncoupling protein 3 protects aconitase against inactivation in isolated skeletal muscle mitochondria. Biochim. Biophys. Acta 1709, 150–156
    1. Tanchot C., Terme M., Pere H., Tran T., Benhamouda N., Strioga M., et al. (2013). Tumor-infiltrating regulatory T cells: phenotype, role, mechanism of expansion in situ and clinical significance. Cancer Microenviron. 6, 147–157 10.1007/s12307-012-0122-y
    1. Teng M. W., Swann J. B., Koebel C. M., Schreiber R. D., Smyth M. J. (2008). Immune-mediated dormancy: an equilibrium with cancer. J. Leukoc. Biol. 84, 988–993 10.1189/jlb.1107774
    1. Tolon R. M., Sanchez-Franco F., Lopez Fernandez J., Lorenzo M. J., Vazquez G. F., Cacicedo L. (1996). Regulation of somatostatin gene expression by veratridine-induced depolarization in cultured fetal cerebrocortical cells. Brain Res. Mol. Brain Res. 35, 103–110 10.1016/0169-328X(95)00188-X
    1. Vanan I., Dong Z., Tosti E., Warshaw G., Symons M., Ruggieri R. (2012). Role of a DNA damage checkpoint pathway in ionizing radiation-induced glioblastoma cell migration and invasion. Cell Mol. Neurobiol. 10.1007/s10571-012-9846-y
    1. Veiseh M., Gabikian P., Bahrami S. B., Veiseh O., Zhang M., Hackman R. C., et al. (2007). Tumor paint: a chlorotoxin:Cy5.5 bioconjugate for intraoperative visualization of cancer foci. Cancer Res. 67, 6882–6888 10.1158/0008-5472.CAN-06-3948
    1. Verheij M. (2008). Clinical biomarkers and imaging for radiotherapy-induced cell death. Cancer Metastasis Rev. 27, 471–480 10.1007/s10555-008-9131-1
    1. Vianello F., Papeta N., Chen T., Kraft P., White N., Hart W. K., et al. (2006). Murine B16 melanomas expressing high levels of the chemokine stromal-derived factor-1/CXCL12 induce tumor-specific T cell chemorepulsion and escape from immune control. J. Immunol. 176, 2902–2914
    1. Vidal-Puig A. J., Grujic D., Zhang C. Y., Hagen T., Boss O., Ido Y., et al. (2000). Energy metabolism in uncoupling protein 3 gene knockout mice. J. Biol. Chem. 275, 16258–16266 10.1074/jbc.M910179199
    1. Voehringer D. W., Hirschberg D. L., Xiao J., Lu Q., Roederer M., Lock C. B., et al. (2000). Gene microarray identification of redox and mitochondrial elements that control resistance or sensitivity to apoptosis. Proc. Natl. Acad. Sci. U.S.A. 97, 2680–2685 10.1073/pnas.97.6.2680
    1. Watanabe H., Bohensky J., Freeman T., Srinivas V., Shapiro I. M. (2008). Hypoxic induction of UCP3 in the growth plate: UCP3 suppresses chondrocyte autophagy. J. Cell Physiol. 216, 419–425 10.1002/jcp.21408
    1. Watkins S., Sontheimer H. (2011). Hydrodynamic cellular volume changes enable glioma cell invasion. J. Neurosci. 31, 17250–17259 10.1523/JNEUROSCI.3938-11.2011
    1. Weaver A. K., Bomben V. C., Sontheimer H. (2006). Expression and function of calcium-activated potassium channels in human glioma cells. Glia 54, 223–233 10.1002/glia.20364
    1. Weber D. C., Casanova N., Zilli T., Buchegger F., Rouzaud M., Nouet P., et al. (2009). Recurrence pattern after [(18)F]fluoroethyltyrosine-positron emission tomography-guided radiotherapy for high-grade glioma: a prospective study. Radiother. Oncol. 93, 586–592 10.1016/j.radonc.2009.08.043
    1. Weihua Z., Tsan R., Huang W. C., Wu Q., Chiu C. H., Fidler I. J., et al. (2008). Survival of cancer cells is maintained by EGFR independent of its kinase activity. Cancer Cell 13, 385–393 10.1016/j.ccr.2008.03.015
    1. Wick W., Wick A., Schulz J. B., Dichgans J., Rodemann H. P., Weller M. (2002). Prevention of irradiation-induced glioma cell invasion by temozolomide involves caspase 3 activity and cleavage of focal adhesion kinase. Cancer Res. 62, 1915–1919
    1. Wild-Bode C., Weller M., Rimner A., Dichgans J., Wick W. (2001). Sublethal irradiation promotes migration and invasiveness of glioma cells: implications for radiotherapy of human glioblastoma. Cancer Res. 61, 2744–2750
    1. Withers H. R. (Ed). (1975). “The four R's of radiotherapy,” in Advances in Radiation Biology, Vol. 5, ed Lett J. T. A. H. (New York, NY: Academic Press; ).
    1. Wright E. M., Loo D. D., Hirayama B. A. (2011). Biology of human sodium glucose transporters. Physiol. Rev. 91, 733–794 10.1152/physrev.00055.2009
    1. Wulff H., Castle N. A. (2010). Therapeutic potential of KCa3.1 blockers: recent advances and promising trends. Expert Rev. Clin. Pharmacol. 3, 385–396 10.1586/ecp.10.11
    1. Yellon D. M., Hausenloy D. J. (2007). Myocardial reperfusion injury. N. Engl. J. Med. 357, 1121–1135 10.1056/NEJMra071667
    1. Yu L. C., Huang C. Y., Kuo W. T., Sayer H., Turner J. R., Buret A. G. (2008). SGLT-1-mediated glucose uptake protects human intestinal epithelial cells against Giardia duodenalis-induced apoptosis. Int. J. Parasitol. 38, 923–934 10.1016/j.ijpara.2007.12.004
    1. Zhao J., Du C. Z., Sun Y. S., Gu J. (2012). Patterns and prognosis of locally recurrent rectal cancer following multidisciplinary treatment. World J. Gastroenterol. 18, 7015–7020 10.3748/wjg.v18.i47.7015
    1. Zhau H. E., He H., Wang C. Y., Zayzafoon M., Morrissey C., Vessella R. L., et al. (2011). Human prostate cancer harbors the stem cell properties of bone marrow mesenchymal stem cells. Clin. Cancer Res. 17, 2159–2169 10.1158/1078-0432.CCR-10-2523
    1. Zhou Y. C., Liu J. Y., Li J., Zhang J., Xu Y. Q., Zhang H. W., et al. (2011). Ionizing radiation promotes migration and invasion of cancer cells through transforming growth factor-beta-mediated epithelial-mesenchymal transition. Int. J. Radiat. Oncol. Biol. Phys. 81, 1530–1537 10.1016/j.ijrobp.2011.06.1956

Source: PubMed

3
S'abonner