DOI QR코드

DOI QR Code

2-deoxy-D-Glucose Synergizes with Doxorubicin or L-Buthionine Sulfoximine to Reduce Adhesion and Migration of Breast Cancer Cells

  • Mustafa, Ebtihal H (Department of Physiology and Biochemistry, Faculty of Medicine, The University of Jordan) ;
  • Mahmoud, Huda T (Department of Biology and Biotechnology, Faculty of Science, The Hashemite University) ;
  • Al-Hudhud, Mariam Y (Department of Physiology and Biochemistry, Faculty of Medicine, The University of Jordan) ;
  • Abdalla, Maher Y (Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center) ;
  • Ahmad, Iman M (Division of Radiation Science Technology Education, University of Nebraska Medical Center) ;
  • Yasin, Salem R (Department of Biology and Biotechnology, Faculty of Science, The Hashemite University) ;
  • Elkarmi, Ali Z (Department of Biology and Biotechnology, Faculty of Science, The Hashemite University) ;
  • Tahtamouni, Lubna H (Department of Biology and Biotechnology, Faculty of Science, The Hashemite University)
  • Published : 2015.04.29

Abstract

Background: Cancer metastasis depends on cell motility which is driven by cycles of actin polymerization and depolymerization. Reactive oxygen species (ROS) and metabolic oxidative stress have long been associated with cancer. ROS play a vital role in regulating actin dynamics that are sensitive to oxidative modification. The current work aimed at studying the effects of sub-lethal metabolic oxidative stress on actin cytoskeleton, focal adhesion and cell migration. Materials and Methods: T47D human breast cancer cells were treated with 2-deoxy-D-glucose (2DG), L-buthionine sulfoximine (BSO), or doxorubicin (DOX), individually or in combination, and changes in intracellular total glutathione and malondialdehyde (MDA) levels were measured. The expression of three major antioxidant enzymes was studied by immunoblotting, and cells were stained with fluorescent-phalloidin to evaluate changes in F-actin organization. In addition, cell adhesion and degradation ability were measured. Cell migration was studied using wound healing and transwell migration assays. Results: Our results show that treating T47D human breast cancer cells with drug combinations (2DG/BSO, 2DG/DOX, or BSO/DOX) decreased intracellular total glutathione and increased oxidized glutathione, lipid peroxidation, and cytotoxicity. In addition, the drug combinations caused a reduction in cell area and mitotic index, prophase arrest and a decreased ability to form invadopodia. The formation of F-actin aggregates was increased in treated T47D cells. Moreover, combination therapy reduced cell adhesion and the rate of cell migration. Conclusions: Our results suggest that exposure of T47D breast cancer cells to combination therapy reduces cell migration via effects on metabolic oxidative stress.

Keywords

References

  1. American Cancer Society. (2014). Breast cancer: detailed guide.
  2. Ahmad I, Mustafa E, Mustafa N, et al (2010). 2DG enhances the susceptibility of breast cancer cells to doxorubicin. Cen Eur J Biol, 5, 739-48.
  3. Ahmad I, Aykin-Burns N, Sim J, et al (2005). Mitochondrial and $H_2O_2$ mediate glucose deprivation-induced stress in human cancer cells. J Biol Chem, 280, 4254-63. https://doi.org/10.1074/jbc.M411662200
  4. Alexandrova A, Kopnin P, Vasiliev J, et al (2006). ROS upregulation mediates Ras-induced changes of cell morphology and motility. Exp Cell Res, 312, 2066-73. https://doi.org/10.1016/j.yexcr.2006.03.004
  5. Anderson E (1985). Determination of glutathione and glutathione disulfide in biological samples. Methods Enzymol, 113, 548-55. https://doi.org/10.1016/S0076-6879(85)13073-9
  6. Andringa K, Coleman M, Aykin-Burns N, et al (2006). Inhibition of glutamate cysteine ligase activity sensitizes human breast cancer cells to the toxicity of 2-deoxy-D-glucose. Cancer Res, 66, 1605-10. https://doi.org/10.1158/0008-5472.CAN-05-3462
  7. Artym V, Yamada K, Mueller S (2009). ECM degradation assays for analyzing local cell invasion. Methods Mol Biol, 522, 211-9. https://doi.org/10.1007/978-1-59745-413-1_15
  8. Averill-Bates D, Przybytkowski E (1994). The role of glucose in cellular defences against cytotoxicity of hydrogen peroxide in Chinese hamster ovary cells. Arch Biochem Biophys, 312, 52-8. https://doi.org/10.1006/abbi.1994.1279
  9. Barth B, Stewart-Smeets S, Kuhn T (2009). Proinflammatory cytokines provoke oxidative damage to actin in neuronal cells mediated by Rac1 and NADPH oxidase. Mol Cell Neurosci, 41, 274-85. https://doi.org/10.1016/j.mcn.2009.03.007
  10. Bertazzoli C, Bellini O, Magrini U et al (1979). Quantitative experimental evaluation of adriamycin cardiotoxicity in the mouse. Cancer Treat Reports, 63, 1877-83.
  11. Chen Q, Vazquez E, Moghaddas S, et al (2003). Production of reactive oxygen species by mitochondria: central role of complex III. J Biol Chem, 278, 36027-31. https://doi.org/10.1074/jbc.M304854200
  12. Coleman M, Asbury C, Daniels D, et al (2008). 2-deoxy-D-glucose causes cytotoxicity, oxidative stress, and radiosensitization in pancreatic cancer. Free Radic Biol Med, 44, 322-31. https://doi.org/10.1016/j.freeradbiomed.2007.08.032
  13. Dong F, Budhu A, Wang X (2009). Translating the metastasis paradigm from scientific theory to clinical oncology. Clin Cancer Res, 15, 2588-93. https://doi.org/10.1158/1078-0432.CCR-08-2356
  14. Estornes Y, Gay F, Gevrey J, et al (2007). Differential involvement of destrin and cofilin-1 in the control of invasive properties of Isreco1 human colon cancer cells. Int J Cancer, 121, 2162-71. https://doi.org/10.1002/ijc.22911
  15. Gavino V, Miller J, Ikharebha S, et al (1981). Effect of polyunsaturated fatty acids and antioxidants on lipid peroxidation in tissue cultures. J Lipid Res, 22, 763-9.
  16. Gille L, Nohl H (1997). Analyses of the molecular mechanism of adriamycin-induced cardiotoxicity. Free Radic Biol Med, 23, 775-82. https://doi.org/10.1016/S0891-5849(97)00025-7
  17. Griffith O (1980). Determination of glutathione and glutathione disulfide using glutathione reductase and 2-vinylpyridine. Anal Biochem, 106, 207-12. https://doi.org/10.1016/0003-2697(80)90139-6
  18. Gupta G, Massague J (2006). Cancer metastasis: building a framework. Cell, 127, 679-95. https://doi.org/10.1016/j.cell.2006.11.001
  19. Gurudath S, Ganapathy K, Pai A, et al (2012). Estimation of superoxide dismutase and glutathione peroxidase in oral submucous fibrosis, oral leukoplakia and oral cancer--a comparative study. Asian Pac J Cancer Prev, 13, 4409-12. https://doi.org/10.7314/APJCP.2012.13.9.4409
  20. Hayot C, Debeir O, Van Ham P, et al (2006). Characterization of the activities of actin-affecting drugs on tumor cell migration. Toxicol Appl Pharmacol, 211, 30-40. https://doi.org/10.1016/j.taap.2005.06.006
  21. Hegde M, Mali A, Chandorkar S (2013). What is a cancer cell? Why does it metastasize? Asian Pac J Cancer Prev, 14, 3987-9. https://doi.org/10.7314/APJCP.2013.14.6.3987
  22. Heng Y, Koh C (2010). Actin cytoskeleton dynamics and the cell division cycle. Int J Biochem Cell Biol, 42, 1622-33. https://doi.org/10.1016/j.biocel.2010.04.007
  23. Jin L, Wei C (2014). Role of microRNAs in the Warburg effect and mitochondrial metabolism in cancer. Asian Pac J Cancer Prev, 15, 7015-9. https://doi.org/10.7314/APJCP.2014.15.17.7015
  24. Korb T, Schluter K, Enns A, et al (2004). Integrity of actin fibers and microtubules influences metastatic tumor cell adhesion. Exp Cell Res, 299, 236-247. https://doi.org/10.1016/j.yexcr.2004.06.001
  25. Lassing I, Schmitzberger F, Bjornstedt M, et al (2007). Molecular and structural basis for redox regulation of beta-actin. J Mol Biol, 370, 331-48. https://doi.org/10.1016/j.jmb.2007.04.056
  26. Lee D, Kang S (2013). Reactive oxygen species and tumor metastasis. Mol Cells, 35, 93-8. https://doi.org/10.1007/s10059-013-0034-9
  27. Lee Y, Galoforo S, Berns C , et al (1998). Glucose deprivationinduced cytotoxicity and alterations in mitogen-activated protein kinase activation are mediated by oxidative stress in multidrug-resistant human breast carcinoma cells. J Biol Chem, 273, 5294-9. https://doi.org/10.1074/jbc.273.9.5294
  28. Li X, Kong X, Wang Y, et al (2013). BRCC2 inhibits breast cancer cell growth and metastasis in vitro and in vivo via downregulating AKT pathway. Cell Death Dis, 4, 757. https://doi.org/10.1038/cddis.2013.290
  29. Lin X, Zhang F, Bradbury C, et al (2003). 2-deoxy-D-glucoseinduced cytotoxicity and radiosensitization in tumor cells is mediated via disruptions in thiol metabolism. Cancer Res, 63, 3413-7.
  30. Luanpitpong S, Talbott S, Rojanasakul Y, et al (2010). Regulation of lung cancer cell migration and invasion by reactive oxygen species and caveolin-1. J Biol Chem, 285, 38832-40. https://doi.org/10.1074/jbc.M110.124958
  31. Lykkesfeldt J (2007). Malondialdehyde as biomarker of oxidative damage to lipids caused by smoking. Clinica Chimica Acta, 380, 50-8. https://doi.org/10.1016/j.cca.2007.01.028
  32. Green M, Graham M, O'Donovan R, et al (2006). Subcellular compartmentalization of glutathione: Correlations with parameters of oxidative stress related to genotoxicity. Mutagenesis, 21, 383-90. https://doi.org/10.1093/mutage/gel043
  33. Maschek G, Savaraj N, Priebe W, et al (2004). 2-deoxy-D-glucose increases the efficacy of adriamycin and paclitaxel in human osteosarcoma and non-small cell lung cancers in vivo. Cancer Res, 64, 31-4. https://doi.org/10.1158/0008-5472.CAN-03-3294
  34. Meng X, Yue S (2014). Dexamethasone disrupts cytoskeleton organization and migration of T47D human breast cancer cells by modulating the AKT/mTOR/RhoA pathway. Asian Pac J Cancer Prev, 15, 10245-50.
  35. Menhofer M, Kubisch R, Schreiner L, et al (2014). The actin targeting compound chondramide inhibits breast cancer metastasis via reduction of cellular contractility. PLoS ONE, 9, 112542. https://doi.org/10.1371/journal.pone.0112542
  36. Meredith M, Reed D (1982). Status of the mitochondrial pool of glutathione in the isolated hepatocyte. J Biol Chem, 257, 3747-53.
  37. Moldovan L, Mythreye K, Goldschmidt-Clermont P, et al (2006). Reactive oxygen species in vascular endothelial cell motility. Roles of NAD(P)H oxidase and Rac1. Cardiovasc Res, 71, 236-46. https://doi.org/10.1016/j.cardiores.2006.05.003
  38. Montanez E, Piwko-Czuchra A, Bauer M, et al (2007). Integrin. (2nd edition ed.). Academic Press, San Diego.
  39. Mosmann T (1983). Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J Immunol Methods, 65, 55-63. https://doi.org/10.1016/0022-1759(83)90303-4
  40. Mu X, Shi W, Sun L, et al (2012). Pristimerin inhibits breast cancer cell migration by up- regulating regulator of G protein signaling 4 expression. Asian Pac J Cancer Prev, 13, 1097-104. https://doi.org/10.7314/APJCP.2012.13.4.1097
  41. Muyderman H, Nilsson M, Sims N (2004). Highly selective and prolonged depletion of mitochondrial glutathione in astrocytes markedly increases sensitivity to peroxynitrite. J Neurosci, 24, 8019-28. https://doi.org/10.1523/JNEUROSCI.1103-04.2004
  42. Nakashima R, Paggi M, Pedersen P (1984). Contributions of glycolysis and oxidative phosphorylation to adenosine 5'-triphosphate production in AS-30D hepatoma cells. Cancer Res, 44, 5702-6.
  43. Noda N, Wakasugi H (2001). Cancer and oxidative stress (Vol. 44).
  44. Nourazarian A, Kangari P, Salmaninejad A (2014). Roles of oxidative stress in the development and progression of breast cancer. Asian Pac J Cancer Prev, 15, 4745-51. https://doi.org/10.7314/APJCP.2014.15.12.4745
  45. Oberley L, Buettner G (1979). Role of superoxide dismutase in cancer: a review. Cancer Res, 39, 1141-9.
  46. Ohkawa H, Ohishi N, Yagi K (1979). Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem, 95, 351-8. https://doi.org/10.1016/0003-2697(79)90738-3
  47. Ortega A, Mena S, Estrela J (2010). Oxidative and nitrosative stress in the metastatic microenvironment. Cancers (Basel), 2, 274-304. https://doi.org/10.3390/cancers2020274
  48. Pani, Galeotti T, Chiarugi P (2010). Metastasis: cancer cell's escape from oxidative stress. Cancer Metastasis Rev, 29, 351-78. https://doi.org/10.1007/s10555-010-9225-4
  49. Sakthivel K, Prabhu V, Guruvayoorappan C (2012). WAVEs: a novel and promising weapon in the cancer therapy tool box. Asian Pac J Cancer Prev, 13, 1719-22. https://doi.org/10.7314/APJCP.2012.13.5.1719
  50. Schumacker P (2006). Reactive oxygen species in cancer cells: live by the sword, die by the sword. Cancer Cell, 10, 175-6. https://doi.org/10.1016/j.ccr.2006.08.015
  51. Shinohara M, Shang W, Kubodera M, et al (2007). Nox1 redox signaling mediates oncogenic Ras-induced disruption of stress fibers and focal adhesions by down-regulating Rho. J Biol Chem, 282, 17640-8. https://doi.org/10.1074/jbc.M609450200
  52. Shiu R, Paterson J (1984). Alteration of cell shape, adhesion, and lipid accumulation in human breast cancer cells (T-47D) by human prolactin and growth hormone. Cancer Res, 44, 1178-86.
  53. Sidani M, Wessels D, Mouneimne G, et al (2007). Cofilin determines the migration behavior and turning frequency of metastatic cancer cells. J Cell Biol, 179, 777-91. https://doi.org/10.1083/jcb.200707009
  54. Sies H (1997). Oxidative stress: oxidants and antioxidants. Exp Physiol, 82, 291-5. https://doi.org/10.1113/expphysiol.1997.sp004024
  55. Simons A, Ahmad I, Mattson D, et al (2007). 2-Deoxy-D-glucose combined with cisplatin enhances cytotoxicity via metabolic oxidative stress in human head and neck cancer cells. Cancer Res, 67, 3364-70. https://doi.org/10.1158/0008-5472.CAN-06-3717
  56. Siveski-Iliskovic N, Hill M, Chow D, et al (1995). Probucol protects against adriamycin cardiomyopathy without interfering with its antitumor effect. American heart association. Circulation, 91, 10-5. https://doi.org/10.1161/01.CIR.91.1.10
  57. Sottnik J, Lori J, Rose B, et al (2011). Glycolysis inhibition by 2-deoxy-D-glucose reverts the metastatic phenotype in vitro and in vivo. Clin Exp Metastasis, 28, 865-75. https://doi.org/10.1007/s10585-011-9417-5
  58. Tahtamouni L, Shaw A, Hasan M, et al (2013). Non-overlapping activities of ADF and cofilin-1 during the migration of metastatic breast tumor cells. BMC Cell Biol, 14, 45-61. https://doi.org/10.1186/1471-2121-14-45
  59. Takanashi S, Bachur N (1976). Adriamycin metabolism in man evidence from urinary metabolites. Drug Metabol Dispos, 4, 79-87.
  60. Toyokuni S, Okamoto K, Yodoi J, et al (1995). Persistent oxidative stress in cancer. FEBS Lett, 358, 1-3. https://doi.org/10.1016/0014-5793(94)01368-B
  61. Wiggan O, Shaw A, DeLuca J, et al (2012). ADF/cofilin regulates actomyosin assembly through competitive inhibition of myosin II binding to F-actin. Develop Cell, 22, 530-43. https://doi.org/10.1016/j.devcel.2011.12.026
  62. Wold L, II N, Ren J (2005). Doxorubicin induces cardiomyocyte dysfunction via a p38 MAP kinase-dependent oxidative stress mechanism. Cancer Detect Prev, 29, 294-9. https://doi.org/10.1016/j.cdp.2004.07.008
  63. Wu D, Cederbaum A (2003). Alcohol, oxidative stress, and free radical damage. Alcohol Res Health, 27, 277-84.
  64. Zaidel-Bar R, Cohen M, Addadi L, Geiger B (2004). Hierarchical assembly of cell-matrix adhesion complexes. Biochem Soc Trans, 32, 416-20. https://doi.org/10.1042/bst0320416
  65. Zebda N, Bernard O, Bailly M, et al (2000). Phosphorylation of ADF/cofilin abolishes EGF-induced actin nucleation at the leading edge and subsequent lamellipod extension. J Cell Biol, 151, 1119-28. https://doi.org/10.1083/jcb.151.5.1119
  66. Zhang J, Rubio V, Zheng S, et al (2009). Knockdown of OLA1, a regulator of oxidative stress response, inhibits motility and invasion of breast cancer cells. J Zhejiang Univ Sci B, 10, 796-804. https://doi.org/10.1631/jzus.B0910009
  67. Zhao Y, Butler E, Tan M (2013). Targeting cellular metabolism to improve cancer therapeutics. Cell Death Dis, 4, 532. https://doi.org/10.1038/cddis.2013.60
  68. Zhou S, Starkov A, Froberg M, et al (2001). Cumulative and irreversible cardiac mitochondrial dysfunction induced by doxorubicin. Cancer Res, 61, 771-7.

Cited by

  1. Combined Treatment with 2-Deoxy-D-Glucose and Doxorubicin Enhances the in Vitro Efficiency of Breast Cancer Radiotherapy vol.16, pp.18, 2016, https://doi.org/10.7314/APJCP.2015.16.18.8431
  2. Golgi apparatus dis- and reorganizations studied with the aid of 2-deoxy-d-glucose and visualized by 3D-electron tomography vol.147, pp.4, 2017, https://doi.org/10.1007/s00418-016-1515-7
  3. Establishment of 2D Cell Cultures Derived From 3D MCF-7 Spheroids Displaying a Doxorubicin Resistant Profile pp.18606768, 2018, https://doi.org/10.1002/biot.201800268