全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
ISRN Oncology  2012 

Oxidative Stress and Lipid Peroxidation Products in Cancer Progression and Therapy

DOI: 10.5402/2012/137289

Full-Text   Cite this paper   Add to My Lib

Abstract:

The generation of reactive oxygen species (ROS) and an altered redox status are common biochemical aspects in cancer cells. ROS can react with the polyunsaturated fatty acids of lipid membranes and induce lipid peroxidation. The end products of lipid peroxidation, 4-hydroxynonenal (HNE), have been considered to be a second messenger of oxidative stress. Beyond ROS involvement in carcinogenesis, increased ROS level can inhibit tumor cell growth. Indeed, in tumors in advanced stages, a further increase of oxidative stress, such as that occurs when using several anticancer drugs and radiation therapy, can overcome the antioxidant defenses of cancer cells and drive them to apoptosis. High concentrations of HNE can also induce apoptosis in cancer cells. However, some cells escape the apoptosis induced by chemical or radiation therapy through the adaptation to intrinsic oxidative stress which confers drug resistance. This paper is focused on recent advances in the studies of the relation between oxidative stress, lipid peroxidation products, and cancer progression with particular attention to the pro-oxidant anticancer agents and the drug-resistant mechanisms, which could be modulated to obtain a better response to cancer therapy. 1. Oxidative Stress and Lipid Peroxidation A body of evidence suggests that oxidative stress and resulting lipid peroxidation are involved in various and numerous pathological states including inflammation, atherosclerosis, neurodegenerative diseases, and cancer. The term “oxidative stress” is frequently used to describe the imbalances in redox couples such as those reduced to oxidized glutathione (GSH/GSSG) or NADPH/NADP+ ratios. Such metabolic disturbances not only involve the overproduction of reactive free radicals but also occur via a nonfree radical pathway, for example, by hydrogen peroxide [1]. In such cases, the products of its action are molecules that are enriched in one or more oxygen atoms that are generally considered to be markers of oxidative stress [2]. Reactive oxygen species (ROS) are thought to be the major ones responsible for the alteration of macromolecules which is often termed oxidative stress. ROS are generated as by-products of cellular metabolism, primarily in the mitochondria [3] and include free radicals such as superoxide anion ( ), perhydroxyl radical ( ), hydroxyl radical (?OH), nitric oxide (NO), and other species such as hydrogen peroxide (H2O2), singlet oxygen (1O2), hypochlorous acid (HOCl), and peroxynitrite (ONOO?) [4]. The hydroxyl radical ?OH is the most reactive radical that can arise

References

[1]  B. M. Hybertson, B. Gao, S. K. Bose, and J. M. McCord, “Oxidative stress in health and disease: the therapeutic potential of Nrf2 activation,” Molecular Aspects of Medicine, vol. 32, pp. 234–246, 2011.
[2]  B. Lipinski, “Hydroxyl radical and its scavengers in health and disease,” Oxidative Medicine and Cellular Longevity, vol. 2011, Article ID 809696, 9 pages, 2011.
[3]  M. L. Riess, A. K. S. Camara, L. G. Kevin, J. An, and D. F. Stowe, “Reduced reactive O2 species formation and preserved mitochondrial NADH and [Ca2+] levels during short-term 17°C ischemia in intact hearts,” Cardiovascular Research, vol. 61, no. 3, pp. 580–590, 2004.
[4]  J. M. Lü, P. H. Lin, Q. Yao, and C. Chen, “Chemical and molecular mechanisms of antioxidants: experimental approaches and model systems,” Journal of Cellular and Molecular Medicine, vol. 14, no. 4, pp. 840–860, 2010.
[5]  T. A. Dix and J. Aikens, “Mechanisms and biological relevance of lipid peroxidation initiation,” Chemical Research in Toxicology, vol. 6, no. 1, pp. 2–18, 1993.
[6]  Z. Y. Cheng and Y. Z. Li, “What is responsible for the initiating chemistry of iron-mediated lipid peroxidation: an update,” Chemical Reviews, vol. 107, no. 3, pp. 748–766, 2007.
[7]  R. A. Weisiger and I. Fridovich, “Superoxide dismutase. Organelle specificity,” Journal of Biological Chemistry, vol. 248, no. 10, pp. 3582–3592, 1973.
[8]  W. Dr?ge, “Free radicals in the physiological control of cell function,” Physiological Reviews, vol. 82, no. 1, pp. 47–95, 2002.
[9]  M. K. Misra, M. Sarwat, P. Bhakuni, R. Tuteja, and N. Tuteja, “Oxidative stress and ischemic myocardial syndromes,” Medical Science Monitor, vol. 15, no. 10, pp. RA209–RA219, 2009.
[10]  B. Halliwell, S. Chirico, M. A. Crawford, K. S. Bjerve, and K. F. Gey, “Lipid peroxidation: its mechanism, measurement, and significance,” American Journal of Clinical Nutrition, vol. 57, no. 5, pp. 715S–724S, 1993.
[11]  H. W. Gardner, “Oxygen radical chemistry of polyunsaturated fatty acids,” Free Radical Biology and Medicine, vol. 7, no. 1, pp. 65–86, 1989.
[12]  P. Spiteller, W. Kern, J. Reiner, and G. Spiteller, “Aldehydic lipid peroxidation products derived from linoleic acid,” Biochimica et Biophysica Acta, vol. 1531, no. 3, pp. 188–208, 2001.
[13]  G. Barrera, S. Pizzimenti, and M. U. Dianzani, “Lipid peroxidation: control of cell proliferation, cell differentiation and cell death,” Molecular Aspects of Medicine, vol. 29, no. 1-2, pp. 1–8, 2008.
[14]  S. Pizzimenti, C. Toaldo, P. Pettazzoni, M. U. Dianzani, and G. Barrera, “The “Two-Faced” effects of reactive oxygen species and the lipid peroxidation product 4-Hydroxynonenal in the hallmarks of cancer,” Cancers, vol. 2, no. 2, pp. 338–363, 2010.
[15]  H. Esterbauer, R. J. Schaur, and H. Zollner, “Chemistry and Biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes,” Free Radical Biology and Medicine, vol. 11, no. 1, pp. 81–128, 1991.
[16]  K. Uchida, “4-Hydroxy-2-nonenal: a product and mediator of oxidative stress,” Progress in Lipid Research, vol. 42, no. 4, pp. 318–343, 2003.
[17]  K. Uchida, L. I. Szweda, H. Z. Chae, and E. R. Stadtman, “Immunochemical detection of 4-hydroxynonenal protein adducts in oxidized hepatocytes,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 18, pp. 8742–8746, 1993.
[18]  L. M. Sayre, W. Sha, G. Xu et al., “Immunochemical evidence supporting 2-pentylpyrrole formation on proteins exposed to 4-hydroxy-2-nonenal,” Chemical Research in Toxicology, vol. 9, no. 7, pp. 1194–1201, 1996.
[19]  G. Xu, Y. Liu, and L. M. Sayre, “Independent synthesis, solution behavior, and studies on the mechanism of formation of a primary amine-derived fluorophore representing cross- linking of proteins by (E)-4-hydroxy-2-nonenal,” Journal of Organic Chemistry, vol. 64, no. 16, pp. 5732–5745, 1999.
[20]  H. J. Forman, J. M. Fukuto, T. Miller, H. Zhang, A. Rinna, and S. Levy, “The chemistry of cell signaling by reactive oxygen and nitrogen species and 4-hydroxynonenal,” Archives of Biochemistry and Biophysics, vol. 477, no. 2, pp. 183–195, 2008.
[21]  T. P. Szatrowski and C. F. Nathan, “Production of large amounts of hydrogen peroxide by human tumor cells,” Cancer Research, vol. 51, no. 3, pp. 794–798, 1991.
[22]  S. Kawanishi, Y. Hiraku, S. Pinlaor, and N. Ma, “Oxidative and nitrative DNA damage in animals and patients with inflammatory diseases in relation to inflammation-related carcinogenesis,” Biological Chemistry, vol. 387, no. 4, pp. 365–372, 2006.
[23]  Y. Zhou, E. O. Hileman, W. Plunkett, M. J. Keating, and P. Huang, “Free radical stress in chronic lymphocytic leukemia cells and its role in cellular sensitivity to ROS-generating anticancer agents,” Blood, vol. 101, no. 10, pp. 4098–4104, 2003.
[24]  A. S. Kamiguti, L. Serrander, K. Lin et al., “Expression and activity of NOX5 in the circulating malignant B cells of hairy cell leukemia,” Journal of Immunology, vol. 175, no. 12, pp. 8424–8430, 2005.
[25]  E. Chow, C. Thirlwell, F. Macrae, and L. Lipton, “Colorectal cancer and inherited mutations in base-excision repair,” Lancet Oncology, vol. 5, no. 10, pp. 600–606, 2004.
[26]  O. Young, T. Crotty, R. O'Connell, J. O'Sullivan, and A. J. Curran, “Levels of oxidative damage and lipid peroxidation in thyroid neoplasia,” Head and Neck, vol. 32, no. 6, pp. 750–756, 2010.
[27]  B. P. Patel, U. M. Rawal, T. K. Dave et al., “Lipid peroxidation, total antioxidant status, and total thiol levels predict overall survival in patients with oral squamous cell carcinoma,” Integrative Cancer Therapies, vol. 6, no. 4, pp. 365–372, 2007.
[28]  S. M. Tsao, M. C. Yin, and W. H. Liu, “Oxidant stress and B vitamins status in patients with non-small cell lung cancer,” Nutrition and Cancer, vol. 59, no. 1, pp. 8–13, 2007.
[29]  B. Kumar, S. Koul, L. Khandrika, R. B. Meacham, and H. K. Koul, “Oxidative stress is inherent in prostate cancer cells and is required for aggressive phenotype,” Cancer Research, vol. 68, no. 6, pp. 1777–1785, 2008.
[30]  J. Boonstra and J. A. Post, “Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells,” Gene, vol. 337, pp. 1–13, 2004.
[31]  F. Q. Schafer and G. R. Buettner, “Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple,” Free Radical Biology and Medicine, vol. 30, no. 11, pp. 1191–1212, 2001.
[32]  S. Toyokuni, “Persistent oxidative stress in cancer,” FEBS Letters, vol. 358, no. 1, pp. 1–3, 1995.
[33]  T. D. Oberley, “Antioxidant enzyme levels in cancer,” Histology and Histopathology, vol. 12, no. 2, pp. 525–535, 1997.
[34]  Y. Hu, D. G. Rosen, Y. Zhou et al., “Mitochondrial manganese-superoxide dismutase expression in ovarian cancer: role in cell proliferation and response to oxidative stress,” Journal of Biological Chemistry, vol. 280, no. 47, pp. 39485–39492, 2005.
[35]  N. Saydam, A. Kirb, O. Demir et al., “Determination of glutathione, glutathione reductase, glutathione peroxidase and glutathione S-transferase levels in human lung cancer tissues,” Cancer Letters, vol. 119, no. 1, pp. 13–19, 1997.
[36]  Y. Murawaki, H. Tsuchiya, T. Kanbe et al., “Aberrant expression of selenoproteins in the progression of colorectal cancer,” Cancer Letters, vol. 259, no. 2, pp. 218–230, 2008.
[37]  K. Irani, Y. Xia, J. L. Zweier et al., “Mitogenic signaling mediated by oxidants in Ras-transformed fibroblasts,” Science, vol. 275, no. 5306, pp. 1649–1652, 1997.
[38]  M. S. Rodrigues, M. M. Reddy, and M. Sattler, “Cell cycle regulation by oncogenic tyrosine kinases in myeloid neoplasias: from molecular redox mechanisms to health implications,” Antioxidants and Redox Signaling, vol. 10, no. 10, pp. 1813–1848, 2008.
[39]  M. Brandon, P. Baldi, and D. C. Wallace, “Mitochondrial mutations in cancer,” Oncogene, vol. 25, no. 34, pp. 4647–4662, 2006.
[40]  Y. S. Bae, J. Y. Sung, O. S. Kim et al., “Platelet-derived growth factor-induced H2O2 production requires the activation of phosphatidylinositol 3-kinase,” Journal of Biological Chemistry, vol. 275, no. 14, pp. 10527–10531, 2000.
[41]  G. Y. Liou and P. Storz, “Reactive oxygen species in cancer,” Free Radical Research, vol. 44, no. 5, pp. 479–496, 2010.
[42]  F. Balkwill, “Tumour necrosis factor and cancer,” Nature Reviews Cancer, vol. 9, no. 5, pp. 361–371, 2009.
[43]  C. Berasain, J. Castillo, M. J. Perugorria, M. U. Latasa, J. Prieto, and M. A. Avila, “Inflammation and liver cancer: new molecular links,” Annals of the New York Academy of Sciences, vol. 1155, pp. 206–221, 2009.
[44]  A. Hammer, M. Ferro, H. M. Tillian et al., “Effect of oxidative stress by iron on 4-hydroxynonenal formation and proliferative activity in hepatomas of different degrees of differentiation,” Free Radical Biology and Medicine, vol. 23, no. 1, pp. 26–33, 1997.
[45]  R. A. Canuto, G. Muzio, M. Maggiora, M. E. Biocca, and M. U. Dianzani, “Glutathione-S-transferase, alcohol dehydrogenase and aldehyde reductase activities during diethylnitrosamine-carcinogenesis in rat liver,” Cancer Letters, vol. 68, no. 2-3, pp. 177–183, 1993.
[46]  R. B. Tjalkens, L. W. Cook, and D. R. Petersen, “Formation and export of the glutathione conjugate of 4-hydroxy-2,3-E- nonenal (4-HNE) in hepatoma cells,” Archives of Biochemistry and Biophysics, vol. 361, no. 1, pp. 113–119, 1999.
[47]  T. D. Oberley, S. Toyokuni, and L. I. Szweda, “Localization of hydroxynonenal protein adducts in normal human kidney and selected human kidney cancers,” Free Radical Biology and Medicine, vol. 27, no. 5-6, pp. 695–703, 1999.
[48]  D. Zanetti, G. Poli, B. Vizio, B. Zingaro, E. Chiarpotto, and F. Biasi, “4-Hydroxynonenal and transforming growth factor-β1 expression in colon cancer,” Molecular Aspects of Medicine, vol. 24, no. 4-5, pp. 273–280, 2003.
[49]  E. Skrzydlewska, A. Stankiewicz, M. Sulkowska, S. Sulkowski, and I. Kasacka, “Antioxidant status and lipid peroxidation in colorectal cancer,” Journal of Toxicology and Environmental Health A, vol. 64, no. 3, pp. 213–222, 2001.
[50]  O. Young, T. Crotty, R. O'Connell, J. O'Sullivan, and A. J. Curran, “Levels of oxidative damage and lipid peroxidation in thyroid neoplasia,” Head and Neck, vol. 32, no. 6, pp. 750–756, 2010.
[51]  G. Juric-Sekhar, K. Zarkovic, G. Waeg, A. Cipak, and N. Zarkovic, “Distribution of 4-hydroxynonenal-protein conjugates as a marker of lipid peroxidation and parameter of malignancy in astrocytic and ependymal tumors of the brain,” Tumori, vol. 95, no. 6, pp. 762–768, 2009.
[52]  P. Karihtala, S. Kauppila, U. Puistola, and A. Jukkola-Vuorinen, “Divergent behaviour of oxidative stress markers 8-hydroxydeoxyguanosine (8-OHdG) and 4-hydroxy-2-nonenal (HNE) in breast carcinogenesis,” Histopathology, vol. 58, no. 6, pp. 854–862, 2011.
[53]  S. Dwivedi, A. Sharma, B. Patrick, R. Sharma, and Y. C. Awasthi, “Role of 4-hydroxynonenal and its metabolites in signaling,” Redox Report, vol. 12, no. 1-2, pp. 4–10, 2007.
[54]  P. Storz, “Reactive oxygen species in tumor progression,” Frontiers in Bioscience, vol. 10, no. 2, pp. 1881–1896, 2005.
[55]  M. Parola, G. Bellomo, G. Robino, G. Barrera, and M. U. Dianzani, “4-Hydroxynonenal as a biological signal: molecular basis and pathophysiological implications,” Antioxidants & Redox Signaling, vol. 1, no. 3, pp. 255–284, 1999.
[56]  T. Finkel and N. J. Holbrook, “Oxidants, oxidative stress and the biology of ageing,” Nature, vol. 408, no. 6809, pp. 239–247, 2000.
[57]  J. L. Martindale and N. J. Holbrook, “Cellular response to oxidative stress: signaling for suicide and survival,” Journal of Cellular Physiology, vol. 192, no. 1, pp. 1–15, 2002.
[58]  M. Benhar, D. Engelberg, and A. Levitzki, “ROS, stress-activated kinases and stress signaling in cancer,” EMBO Reports, vol. 3, no. 5, pp. 420–425, 2002.
[59]  S. S. Leonard, G. K. Harris, and X. Shi, “Metal-induced oxidative stress and signal transduction,” Free Radical Biology and Medicine, vol. 37, no. 12, pp. 1921–1942, 2004.
[60]  J. Boonstra and J. A. Post, “Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells,” Gene, vol. 337, pp. 1–13, 2004.
[61]  R. C. Rancourt, P. C. Keng, C. E. Helt, and M. A. O'Reilly, “The role of in growth of epithelial cells exposed to hyperoxia,” American Journal of Physiology, vol. 280, no. 4, pp. L617–L626, 2001.
[62]  J. S. Shenberger and P. S. Dixon, “Oxygen induces S-phase growth arrest and increases p53 and p21WAF1/CIP1 expression in human bronchial smooth-muscle cells,” American Journal of Respiratory Cell and Molecular Biology, vol. 21, no. 3, pp. 395–402, 1999.
[63]  C. E. Helt, R. C. Rancourt, R. J. Staversky, and M. A. O'Reilly, “p53-dependent induction of p21Cip1/WAF1/Sdi1 protects against oxygen-induced toxicity,” Toxicological Sciences, vol. 63, no. 2, pp. 214–222, 2001.
[64]  C. Martìnez Mu?oz, L. A. van Meeteren, J. A. Post, A. J. Verkleij, C. T. Verrips, and J. Boonstra, “Hydrogen peroxide inhibits cell cycle progression by inhibition of the spreading of mitotic CHO cells,” Free Radical Biology and Medicine, vol. 33, no. 8, pp. 1061–1072, 2002.
[65]  T. Thorn, R. Gniadecki, A. B. Petersen, J. Vicanova, and H. C. Wulf, “Differences in activation of G2/M checkpoint in keratinocytes after genotoxic stress induced by hydrogen peroxide and ultraviolet a radiation,” Free Radical Research, vol. 35, no. 4, pp. 405–416, 2001.
[66]  Y. W. Chung, D. W. Jeong, J. Yun Won, E. J. Choi, Y. Hyun Choi, and I. Young Kim, “H2O2-induced AP-1 activation and its effect on -mediated G2/M arrest in a p53-deficient human lung cancer cell,” Biochemical and Biophysical Research Communications, vol. 293, no. 4, pp. 1248–1253, 2002.
[67]  P. Zhang, Y. Z. Wang, E. Kagan, and J. C. Bonner, “Peroxynitrite targets the epidermal growth factor receptor, Raf-1, and MEK independently to activate MAPK,” Journal of Biological Chemistry, vol. 275, no. 29, pp. 22479–22486, 2000.
[68]  E. Cadenas, “Mitochondrial free radical production and cell signaling,” Molecular Aspects of Medicine, vol. 25, no. 1-2, pp. 17–26, 2004.
[69]  H. U. Simon, A. Haj-Yehia, and F. Levi-Schaffer, “Role of reactive oxygen species (ROS) in apoptosis induction,” Apoptosis, vol. 5, no. 5, pp. 415–418, 2000.
[70]  Y. M. Chung, Y. S. Bae, and S. Y. Lee, “Molecular ordering of ROS production, mitochondrial changes, and caspase activation during sodium salicylate-induced apoptosis,” Free Radical Biology and Medicine, vol. 34, no. 4, pp. 434–442, 2003.
[71]  M. Caron, M. Auclair, A. Vissian, C. Vigouroux, and J. Capeau, “Contribution of mitochondrial dysfunction and oxidative stress to cellular premature senescence induced by antiretroviral thymidine analogues,” Antiviral Therapy, vol. 13, no. 1, pp. 27–38, 2008.
[72]  K. Ksiazek, J. F. Passos, S. Olijslagers, and T. von Zglinicki, “Mitochondrial dysfunction is a possible cause of accelerated senescence of mesothelial cells exposed to high glucose,” Biochemical and Biophysical Research Communications, vol. 366, no. 3, pp. 793–799, 2008.
[73]  J. F. Passos, T. von Zglinicki, and G. Saretzki, “Mitochondrial dysfunction and cell senescence: cause or consequence?” Rejuvenation Research, vol. 9, no. 1, pp. 64–68, 2006.
[74]  J. W. Shay, O. M. Pereira-Smith, and W. E. Wright, “A role for both RB and p53 in the regulation of human cellular senescence,” Experimental Cell Research, vol. 196, no. 1, pp. 33–39, 1991.
[75]  Z. Chen, L. C. Trotman, D. Shaffer et al., “Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis,” Nature, vol. 436, no. 7051, pp. 725–730, 2005.
[76]  K. S. Kim, K. W. Kang, Y. B. Seu, S. H. Baek, and J. R. Kim, “Interferon-γ induces cellular senescence through p53-dependent DNA damage signaling in human endothelial cells,” Mechanisms of Ageing and Development, vol. 130, no. 3, pp. 179–188, 2009.
[77]  R. C. Allsopp, E. Chang, M. Kashefi-Aazam et al., “Telomere shortening is associated with cell division in vitro and in vivo,” Experimental Cell Research, vol. 220, no. 1, pp. 194–200, 1995.
[78]  S. Oikawa and S. Kawanishi, “Site-specific DNA damage at GGG sequence by oxidative stress may accelerate telomere shortening,” FEBS Letters, vol. 453, no. 3, pp. 365–368, 1999.
[79]  W. C. Chou, H. Y. Chen, S. L. Yu, L. Cheng, P. C. Yang, and C. V. Dang, “Arsenic suppresses gene expression in promyelocytic leukemia cells partly through Sp1 oxidation,” Blood, vol. 106, no. 1, pp. 304–310, 2005.
[80]  G. Barrera, S. Martinotti, V. Fazio et al., “Effect of 4-hydroxynonenal on c-myc expression,” Toxicologic Pathology, vol. 15, no. 2, pp. 238–240, 1987.
[81]  V. M. Fazio, G. Barrera, S. Martinotti et al., “4-Hydroxynonenal, a product of cellular lipid peroxidation, which modulates c-myc and globin gene expression in K562 erythroleukemic cells,” Cancer Research, vol. 52, no. 18, pp. 4866–4871, 1992.
[82]  G. Barrera, R. Muraca, S. Pizzimenti et al., “Inhibition of c-myc expression induced by 4-hydroxynonenal, a product of lipid peroxidation, in the HL-60 human leukemic cell line,” Biochemical and Biophysical Research Communications, vol. 203, no. 1, pp. 553–561, 1994.
[83]  G. Barrera, S. Pizzimenti, A. Serra et al., “4-hydroxynonenal specifically inhibits c-myb but does not affect c-fos expressions in HL-60 cells,” Biochemical and Biophysical Research Communications, vol. 227, no. 2, pp. 589–593, 1996.
[84]  S. Pizzimenti, F. Briatore, S. Laurora et al., “4-Hydroxynonenal inhibits telomerase activity and hTERT expression in human leukemic cell lines,” Free Radical Biology and Medicine, vol. 40, no. 9, pp. 1578–1591, 2006.
[85]  M. Rinaldi, G. Barrera, A. Aquino et al., “4-Hydroxynonenal-induced MEL cell differentiation involves PKC activity translocation,” Biochemical and Biophysical Research Communications, vol. 272, no. 1, pp. 75–80, 2000.
[86]  A. Cerbone, C. Toaldo, S. Laurora et al., “4-Hydroxynonenal and PPARγ ligands affect proliferation, differentiation, and apoptosis in colon cancer cells,” Free Radical Biology and Medicine, vol. 42, no. 11, pp. 1661–1670, 2007.
[87]  G. Barrera, S. Pizzimenti, R. Muraca et al., “Effect of 4-hydroxynonenal on cell cycle progression and expression of differentiation-associated antigens in HL-60 cells,” Free Radical Biology and Medicine, vol. 20, no. 3, pp. 455–462, 1996.
[88]  S. Pizzimenti, G. Barrera, M. U. Dianzani, and S. Brüsselbach, “Inhibition of D1, D2, and A cyclin expression in HL-60 cells by the lipid peroxydation product 4-hydroxynonenal,” Free Radical Biology and Medicine, vol. 26, no. 11-12, pp. 1578–1586, 1999.
[89]  G. Barrera, S. Pizzimenti, S. Laurora, E. Moroni, B. Giglioni, and M. U. Dianzani, “4-Hydroxynonenal affects pRb/E2F pathway in HL-60 human leukemic cells,” Biochemical and Biophysical Research Communications, vol. 295, no. 2, pp. 267–275, 2002.
[90]  G. Barrera, S. Pizzimenti, and M. U. Dianzani, “4-Hydroxynonenal and regulation of cell cycle: effects on the pRb/E2F pathway,” Free Radical Biology and Medicine, vol. 37, no. 5, pp. 597–606, 2004.
[91]  S. Laurora, E. Tamagno, F. Briatore et al., “4-Hydroxynonenal modulation of p53 family gene expression in the SK-N-BE neuroblastoma cell line,” Free Radical Biology and Medicine, vol. 38, no. 2, pp. 215–225, 2005.
[92]  K. Shiraishi and K. Naito, “Effects of 4-hydroxy-2-nonenal, a marker of oxidative stress, on spermatogenesis and expression of p53 protein in male infertility,” Journal of Urology, vol. 178, no. 3, pp. 1012–1017, 2007.
[93]  B. Vizio, G. Poli, E. Chiarpotto, and F. Biasi, “4-Hydroxynonenal and TGF-β1 concur in inducing antiproliferative effects on the CaCo-2 human colon adenocarcinoma cell line,” BioFactors, vol. 24, no. 1–4, pp. 237–246, 2005.
[94]  D. Zanetti, G. Poli, B. Vizio, B. Zingaro, E. Chiarpotto, and F. Biasi, “4-Hydroxynonenal and transforming growth factor-β1 expression in colon cancer,” Molecular Aspects of Medicine, vol. 24, no. 4-5, pp. 273–280, 2003.
[95]  C. D. Albright, E. Klem, A. A. Shah, and P. Gallagher, “Breast cancer cell-targeted oxidative stress: enhancement of cancer cell uptake of conjugated linoleic acid, activation of p53, and inhibition of proliferation,” Experimental and Molecular Pathology, vol. 79, no. 2, pp. 118–125, 2005.
[96]  S. B. Sunjic, A. Cipak, F. Rabuzin, R. Wildburger, and N. Zarkovic, “The influence of 4-hydroxy-2-nonenal on proliferation, differentiation and apoptosis of human osteosarcoma cells,” BioFactors, vol. 24, no. 1–4, pp. 141–148, 2005.
[97]  P. Pettazzoni, S. Pizzimenti, C. Toaldo et al., “Induction of cell cycle arrest and DNA damage by the HDAC inhibitor panobinostat (LBH589) and the lipid peroxidation end product 4-hydroxynonenal in prostate cancer cells,” Free Radical Biology and Medicine, vol. 50, no. 2, pp. 313–322, 2011.
[98]  R. A. Canuto, G. Muzio, M. Ferro et al., “Inhibition of class-3 aldehyde dehydrogenase and cell growth by restored lipid peroxidation in hepatoma cell lines,” Free Radical Biology and Medicine, vol. 26, no. 3-4, pp. 333–340, 1999.
[99]  G. Muzio, A. Trombetta, G. Martinasso, R. A. Canuto, and M. Maggiora, “Antisense oligonucleotides against aldehyde dehydrogenase 3 inhibit hepatoma cell proliferation by affecting MAP kinases,” Chemico-Biological Interactions, vol. 143-144, pp. 37–43, 2003.
[100]  L. Schneider, S. Giordano, B. R. Zelickson, et al., “Differentiation of SH-SY5Y cells to a neuronal phenotype changes cellular bioenergetics and the response to oxidative stress,” Free Radical Biology and Medicine, vol. 51, pp. 2007–2017, 2011.
[101]  H. Kakishita and Y. Hattori, “Vascular smooth muscle cell activation and growth by 4-hydroxynonenal,” Life Sciences, vol. 69, no. 6, pp. 689–697, 2001.
[102]  T. Watanabe, R. Pakala, T. Katagiri, and C. R. Benedict, “Lipid peroxidation product 4-hydroxy-2-nonenal acts synergistically with serotonin in inducing vascular smooth muscle cell proliferation,” Atherosclerosis, vol. 155, no. 1, pp. 37–44, 2001.
[103]  T. J. Lee, J. T. Lee, S. K. Moon, C. H. Kim, J. W. Park, and T. K. Kwon, “Age-related differential growth rate and respo61nse to 4-hydroxynonenal in mouse aortic smooth muscle cells,” International Journal of Molecular Medicine, vol. 17, pp. 29–35, 2006.
[104]  C. Vindis, I. Escargueil-Blanc, K. Uchida, M. Elbaz, R. Salvayre, and A. Negre-Salvayre, “Lipid oxidation products and oxidized low-density lipoproteins impair platelet-derived growth factor receptor activity in smooth muscle cells: implication in atherosclerosis,” Redox Report, vol. 12, no. 1-2, pp. 96–100, 2007.
[105]  T. Semlitsch, H. M. Tillian, N. Zarkovic et al., “Differential influence of the lipid peroxidation product 4-hydroxynonenal on the growth of human lymphatic leukaemia cells and human periopherial blood lymphocytes,” Anticancer Research, vol. 22, no. 3, pp. 1689–1697, 2002.
[106]  D. C. Hassane, M. L. Guzman, C. Corbett et al., “Discovery of agents that eradicate leukemia stem cells using an in silico screen of public gene expression data,” Blood, vol. 111, no. 12, pp. 5654–5662, 2008.
[107]  L. Behrend, G. Henderson, and R. M. Zwacka, “Reactive oxygen species in oncogenic transformation,” Biochemical Society Transactions, vol. 31, no. 6, pp. 1441–1444, 2003.
[108]  W. S. Wu, “The signaling mechanism of ROS in tumor progression,” Cancer and Metastasis Reviews, vol. 25, no. 4, pp. 695–705, 2006.
[109]  H. Pelicano, D. Carney, and P. Huang, “ROS stress in cancer cells and therapeutic implications,” Drug Resistance Updates, vol. 7, no. 2, pp. 97–110, 2004.
[110]  P. T. Schumacker, “Reactive oxygen species in cancer cells: live by the sword, die by the sword,” Cancer Cell, vol. 10, no. 3, pp. 175–176, 2006.
[111]  E. J. Wolvetang, K. L. Johnson, K. Krauer, S. J. Ralph, and A. W. Linnane, “.Mitochondrial respiratory chain inhibitors induce apoptosis,” FEBS Letters, vol. 339, pp. 40–44, 1994.
[112]  J. S. Isenberg, K. L. Kolaja, S. A. Ayoubi, J. B. Watkins III, and J. E. Klaunig, “Inhibition of WY-14,643 induced hepatic lesion growth in mice by rotenone,” Carcinogenesis, vol. 18, no. 8, pp. 1511–1519, 1997.
[113]  J. S. Armstrong, B. Hornung, P. Lecane, D. P. Jones, and S. J. Knox, “Rotenone-induced G2/M cell cycle arrest and apoptosis in a human B lymphoma cell line PW,” Biochemical and Biophysical Research Communications, vol. 289, no. 5, pp. 973–978, 2001.
[114]  S. Tada-Oikawa, Y. Hiraku, M. Kawanishi, and S. Kawanishi, “Mechanism for generation of hydrogen peroxide and change of mitochondrial membrane potential during rotenone-induced apoptosis,” Life Sciences, vol. 73, no. 25, pp. 3277–3288, 2003.
[115]  W. G. Chung, C. L. Miranda, and C. S. Maier, “Epigallocatechin gallate (EGCG) potentiates the cytotoxicity of rotenone in neuroblastoma SH-SY5Y cells,” Brain Research, vol. 1176, no. 1, pp. 133–142, 2007.
[116]  Y. T. Deng, H. C. Huang, and J. K. Lin, “Rotenone induces apoptosis in MCF-7 human breast cancer cell-mediated ROS through JNK and p38 signaling,” Molecular Carcinogenesis, vol. 49, no. 2, pp. 141–151, 2010.
[117]  M. T. Hoyt, R. Palchaudhuri, and P. J. Hergenrother, “Cribrostatin 6 induces death in cancer cells through a reactive oxygen species (ROS)-mediated mechanism,” Investigational New Drugs, vol. 29, no. 4, pp. 562–573, 2011.
[118]  R. Sharma, A. Sharma, P. Chaudhary et al., “Role of lipid peroxidation in cellular responses to d, l -sulforaphane, a promising cancer chemopreventive agent,” Biochemistry, vol. 49, no. 14, pp. 3191–3202, 2010.
[119]  H. J. Wang, Y. Y. Jiang, X. F. Wei et al., “Silibinin induces protective superoxide generation in human breast cancer MCF-7 cells,” Free Radical Research, vol. 44, no. 1, pp. 90–100, 2010.
[120]  W. L. Wu, W. L. Chang, and C. F. Chen, “Cytotoxic activities of tanshinones against human carcinoma cell lines,” American Journal of Chinese Medicine, vol. 19, no. 3-4, pp. 207–216, 1991.
[121]  W. Y. W. Lee, K. W. K. Liu, and J. H. K. Yeung, “Reactive oxygen species-mediated kinase activation by dihydrotanshinone in tanshinones-induced apoptosis in HepG2 cells,” Cancer Letters, vol. 285, no. 1, pp. 46–57, 2009.
[122]  C. Hoelzl, J. Bichler, F. Ferk et al., “Methods for the detection of antioxidants which prevent age related diseases: a critical review with particular emphasis on human intervention studies,” Journal of Physiology and Pharmacology, vol. 56, no. 2, pp. 49–64, 2005.
[123]  L. H. Russell Jr., E. Mazzio, R. B. Badisa, et al., “Autoxidation of gallic acid induces ROS-dependent death in human prostate cancer LNCaP cells,” Anticancer Research, vol. 32, pp. 1595–1602, 2012.
[124]  T. Hongo, N. Miyano-Kurosaki, K. Kurosaki, A. Hata, S. Harigae, and A. Tomoda, “2-aminophenoxazine-3-one prevents pulmonary metastasis of mouse B16 melanoma cells in mice,” Journal of Pharmacological Sciences, vol. 114, no. 1, pp. 63–68, 2010.
[125]  C. L. Zheng, X. F. Che, S. I. Akiyama, K. Miyazawa, and A. Tomoda, “2-Aminophenoxazine-3-one induces cellular apoptosis by causing rapid intracellular acidification and generating reactive oxygen species in human lung adenocarcinoma cells,” International Journal of Oncology, vol. 36, no. 3, pp. 641–650, 2010.
[126]  R. Feng, H. M. Ni, S. Y. Wang et al., “Cyanidin-3-rutinoside, a natural polyphenol antioxidant, selectively kills leukemic cells by induction of oxidative stress,” Journal of Biological Chemistry, vol. 282, no. 18, pp. 13468–13476, 2007.
[127]  J. Cvorovic, F. Tramer, M. Granzotto, L. Candussio, G. Decorti, and S. Passamonti, “Oxidative stress-based cytotoxicity of delphinidin and cyanidin in colon cancer cells,” Archives of Biochemistry and Biophysics, vol. 501, no. 1, pp. 151–157, 2010.
[128]  K. Koprowska and M. Czyz, “Molecular mechanisms of parthenolide's action: old drug with a new face,” Postepy Higieny i Medycyny Doswiadczalnej, vol. 64, pp. 100–114, 2010.
[129]  H. Nakshatri, S. E. Rice, and P. Bhat-Nakshatri, “Antitumor agent parthenolide reverses resistance of breast cancer cells to tumor necrosis factor-related apoptosis-inducing ligand through sustained activation of c-Jun N-terminal kinase,” Oncogene, vol. 23, no. 44, pp. 7330–7344, 2004.
[130]  M. L. Guzman, R. M. Rossi, S. Neelakantan et al., “An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells,” Blood, vol. 110, no. 13, pp. 4427–4435, 2007.
[131]  Y. C. Yang, C. K. Lii, A. H. Lin, et al., “Induction of glutathione synthesis and heme oxygenase 1 by the flavonoids butein and phloretin is mediated through the ERK/Nrf2 pathway and protects against oxidative stress,” Free Radical Biology and Medicine, vol. 51, pp. 2073–2081, 2011.
[132]  H. M. Prince and M. Dickinson, “Romidepsin for cutaneous T-cell lymphoma,” Clinical Cancer Research, vol. 18, no. 13, pp. 3509–3515, 2012.
[133]  H. Mizutani, Y. Hiraku, S. Tada-Oikawa et al., “Romidepsin (FK228), a potent histone deacetylase inhibitor, induces apoptosis through the generation of hydrogen peroxide,” Cancer Science, vol. 101, no. 10, pp. 2214–2219, 2010.
[134]  W. H. Chiu, S. J. Luo, C. L. Chen, et al., “Vinca alkaloids cause aberrant ROS-mediated JNK activation, Mcl-1 downregulation, DNA damage, mitochondrial dysfunction, and apoptosis in lung adenocarcinoma cells,” Biochemical Pharmacology, vol. 83, pp. 1159–1171, 2012.
[135]  C. Casares, R. Ramírez-Camacho, A. Trinidad, A. Roldán, E. Jorge, and J. R. García-Berrocal, “Reactive oxygen species in apoptosis induced by cisplatin: review of physiopathological mechanisms in animal models,” European Archives of Oto-Rhino-Laryngology. In press.
[136]  J. Fang, H. Nakamura, and A. K. Iyer, “Tumor-targeted induction of oxystress for cancer therapy,” Journal of Drug Targeting, vol. 15, no. 7-8, pp. 475–486, 2007.
[137]  K. K. Kim, T. S. Lange, R. K. Singh, L. Brard, and R. G. Moore, “Tetrathiomolybdate sensitizes ovarian cancer cells to anticancer drugs doxorubicin, fenretinide, 5-fluorouracil and mitomycin C,” BMC Cancer, vol. 12, p. 147, 2012.
[138]  Y. W. Zhang, J. Shi, Y. J. Li, and L. Wei, “Cardiomyocyte death in doxorubicin-induced cardiotoxicity,” Archivum Immunologiae et Therapia Experimentalis, vol. 57, pp. 435–445, 2009.
[139]  I. Müller, D. Niethammer, and G. Bruchelt, “Anthracycline-derived chemotherapeutics in apoptosis and free radical cytotoxicity (Review),” International Journal of Molecular Medicine, vol. 1, no. 2, pp. 491–494, 1998.
[140]  Y. Pommier, “DNA topoisomerase I Inhibitors: chemistry, biology, and interfacial inhibition,” Chemical Reviews, vol. 109, no. 7, pp. 2894–2902, 2009.
[141]  Y. Li, C. Rory Goodwin, Y. Sang, E. M. Rosen, J. Laterra, and S. Xia, “Camptothecin and Fas receptor agonists synergistically induce medulloblastoma cell death: ROS-dependent mechanisms,” Anti-Cancer Drugs, vol. 20, no. 9, pp. 770–778, 2009.
[142]  X. J. Wu and X. Hua, “Targeting ROS: selective killing of cancer cells by a cruciferous vegetable derived pro-oxidant compound,” Cancer Biology and Therapy, vol. 6, no. 5, pp. 646–647, 2007.
[143]  X. Wei, W. Guo, S. Wu et al., “Oxidative stress in NSC-741909-induced apoptosis of cancer cells,” Journal of Translational Medicine, vol. 8, article 37, 2010.
[144]  B. Sung, B. Park, V. R. Yadav, and B. B. Aggarwal, “Celastrol, a triterpene, enhances TRAIL-induced apoptosis through the down-regulation of cell survival proteins and up-regulation of death receptors,” Journal of Biological Chemistry, vol. 285, no. 15, pp. 11498–11507, 2010.
[145]  A. K. Maiti, “Genetic determinants of oxidative stress-mediated sensitization of drug-resistant cancer cells,” International Journal of Cancer, vol. 130, no. 1, pp. 1–9, 2012.
[146]  R. Kong, G. Jia, Z. X. Cheng, et al., “Dihydroartemisinin enhances Apo2L/TRAIL-mediated apoptosis in pancreatic cancer cells via ROS-mediated up-regulation of death receptor 5,” PLoS ONE, vol. 7, Article ID e37222, 2012.
[147]  M. A. Park, C. Mitchell, G. Zhang et al., “Vorinostat and sorafenib increase CD95 activation in gastrointestinal tumor cells through a CA2+-de novo ceramide-PP2A-reactive oxygen species-dependent signaling pathway,” Cancer Research, vol. 70, no. 15, pp. 6313–6324, 2010.
[148]  M. Donadelli, C. Costanzo, S. Beghelli et al., “Synergistic inhibition of pancreatic adenocarcinoma cell growth by trichostatin A and gemcitabine,” Biochimica et Biophysica Acta, vol. 1773, no. 7, pp. 1095–1106, 2007.
[149]  H. Maeda, S. Hori, H. Ohizumi et al., “Effective treatment of advanced solid tumors by the combination of arsenic trioxide and L-buthionine-sulfoximine,” Cell Death and Differentiation, vol. 11, no. 7, pp. 737–746, 2004.
[150]  Z. Li, S. Yan, N. Attayan, S. Ramalingam, and C. J. Thiele, “Combination of an allosteric Akt inhibitor MK-2206 with etoposide or rapamycin enhances the antitumor growth effect in neuroblastoma,” Clinical Cancer Research, vol. 18, no. 13, pp. 3603–3615, 2012.
[151]  R. Cuperus, A. B. P. van Kuilenburg, R. Leen, J. Bras, H. N. Caron, and G. A. M. Tytgat, “Promising effects of the 4HPR-BSO combination in neuroblastoma monolayers and spheroids,” Free Radical Biology and Medicine, vol. 51, no. 6, pp. 1213–1220, 2011.
[152]  G. Chen, K. Wang, B. Y. Yang, B. Tang, J. X. Chen, and Z. C. Hua, “Synergistic antitumor activity of oridonin and arsenic trioxide on hepatocellular carcinoma cells,” International Journal of Oncology, vol. 40, pp. 139–147, 2012.
[153]  P. Shen, T. Jiang, H. Lu, H. Han, and R. Luo, “Combination of Poly I: C and arsenic trioxide triggers apoptosis synergistically via activation of TLR3 and mitochondrial pathways in hepatocellular carcinoma cells,” Cell Biology International, vol. 35, pp. 803–810, 2011.
[154]  H. Jiang, Y. Ma, X. Chen et al., “Genistein synergizes with arsenic trioxide to suppress human hepatocellular carcinoma,” Cancer Science, vol. 101, no. 4, pp. 975–983, 2010.
[155]  J. J. Li, Q. Tang, Y. Li et al., “Role of oxidative stress in the apoptosis of hepatocellular carcinoma induced by combination of arsenic trioxide and ascorbic acid,” Acta Pharmacologica Sinica, vol. 27, no. 8, pp. 1078–1084, 2006.
[156]  A. Inoue, S. Muranaka, H. Fujita, T. Kanno, H. Tamai, and K. Utsumi, “Molecular mechanism of diclofenac-induced apoptosis of promyelocytic leukemia: dependency on reactive oxygen species, Akt, Bid, cytochrome c, and caspase pathway,” Free Radical Biology and Medicine, vol. 37, no. 8, pp. 1290–1299, 2004.
[157]  M. Rahmani, E. Reese, Y. Dai et al., “Coadministration of histone deacetylase inhibitors and perifosine synergistically induces apoptosis in human leukemia cells through Akt and ERK1/2 inactivation and the generation of ceramide and reactive oxygen species,” Cancer Research, vol. 65, no. 6, pp. 2422–2432, 2005.
[158]  C. Yu, B. B. Friday, J. P. Lai et al., “Abrogation of MAPK and Akt signaling by AEE788 synergistically potentiates histone deacetylase inhibitor-induced apoptosis through reactive oxygen species generation,” Clinical Cancer Research, vol. 13, no. 4, pp. 1140–1148, 2007.
[159]  F. L. Khanim, R. E. Hayden, J. Birtwistle et al., “Combined bezafibrate and medroxyprogesterone acetate: potential novel therapy for acute myeloid leukaemia,” PLoS ONE, vol. 4, no. 12, Article ID e8147, 2009.
[160]  Y. Jin, Z. Lu, K. Ding et al., “Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: inactivation of the NF-κB pathway and generation of reactive oxygen species,” Cancer Research, vol. 70, no. 6, pp. 2516–2527, 2010.
[161]  S. H. Tonino, J. Van Laar, M. H. Van Oers, J. Y. Wang, E. Eldering, and A. P. Kater, “ROS-mediated upregulation of Noxa overcomes chemoresistance in chronic lymphocytic leukemia,” Oncogene, vol. 30, no. 6, pp. 701–713, 2011.
[162]  S. Sturlan, M. Baumgartner, E. Roth, and T. Bachleitner-Hofmann, “Docosahexaenoic acid enhances arsenic trioxide-mediated apoptosis in arsenic trioxide-resistant HL-60 cells,” Blood, vol. 101, no. 12, pp. 4990–4997, 2003.
[163]  M. Baumgartner, S. Sturlan, E. Roth, B. Wessner, and T. Bachleitner-Hofmann, “Enhancement of arsenic trioxide-mediated apoptosis using docosahexaenoic acid in arsenic trioxide-resistant solid tumor cells,” International Journal of Cancer, vol. 112, no. 4, pp. 707–712, 2004.
[164]  M. Wirtitsch, E. Roth, T. Bachleitner-Hofmann, B. Wessner, and S. Sturlan, “ω-3 and ω-6 polyunsaturated fatty acids enhance arsenic trioxide efficacy in arsenic trioxide-resistant leukemic and solid tumor cells,” Oncology Research, vol. 18, no. 2-3, pp. 83–94, 2009.
[165]  F. M. Santandreu, A. Valle, J. Oliver, and P. Roca, “Resveratrol potentiates the cytotoxic oxidative stress induced by chemotherapy in human colon cancer cells,” Cellular Physiology and Biochemistry, vol. 28, pp. 219–228, 2011.
[166]  F. Cai, Y. M. Dupertuis, and C. Pichard, “Role of polyunsaturated fatty acids and lipid peroxidation on colorectal cancer risk and treatments,” Current Opinion in Clinical Nutrition & Metabolic Care, vol. 15, pp. 99–106, 2012.
[167]  P. Pettazzoni, S. Pizzimenti, C. Toaldo et al., “Induction of cell cycle arrest and DNA damage by the HDAC inhibitor panobinostat (LBH589) and the lipid peroxidation end product 4-hydroxynonenal in prostate cancer cells,” Free Radical Biology and Medicine, vol. 50, no. 2, pp. 313–322, 2011.
[168]  S. Pizzimenti, S. Laurora, F. Briatore, C. Ferretti, M. U. Dianzani, and G. Barrera, “Synergistic effect of 4-hydroxynonenal and PPAR ligands in controlling human leukemic cell growth and differentiation,” Free Radical Biology and Medicine, vol. 32, no. 3, pp. 233–245, 2002.
[169]  B. D. Lawenda, K. M. Kelly, E. J. Ladas, S. M. Sagar, A. Vickers, and J. B. Blumberg, “Should supplemental antioxidant administration be avoided during chemotherapy and radiation therapy?” Journal of the National Cancer Institute, vol. 100, no. 11, pp. 773–783, 2008.
[170]  C. Borek, “Dietary antioxidants and human cancer,” Integrative Cancer Therapies, vol. 3, no. 4, pp. 333–341, 2004.
[171]  B. Halliwell and J. M. C. Gutteridge, Free Radical Biology and Medicine, Oxford Calendron Press, 1989.
[172]  B. N. Pandey and K. P. Mishra, “In-vitro studies on radiation induced membrane oxidative damage in apoptotic death thymocytes,” International Journal of Low Radiation, vol. 1, pp. 113–119, 2003.
[173]  V. Bauer and F. Bauer, “Reactive oxygen species mediators of tissue protection and injury,” General Physiology and Biophysics, vol. 18, pp. 7–14, 1999.
[174]  G. M. Ross, “Induction of cell death by radiotherapy,” Endocrine-Related Cancer, vol. 6, no. 1, pp. 41–44, 1999.
[175]  V. Cardile, M. Bellia, L. Lombardo, C. Scifo, and M. Renis, “Distinct response to ionizing radiation of human prostate cell lines,” Oncology Reports, vol. 14, no. 4, pp. 981–985, 2005.
[176]  K. S. Kumar, M. Raghavan, K. Hieber et al., “Preferential radiation sensitization of prostate cancer in nude mice by nutraceutical antioxidant γ-tocotrienol,” Life Sciences, vol. 78, no. 18, pp. 2099–2104, 2006.
[177]  K. D. Kikawa, J. S. Herrick, R. E. Tateo, M. Mouradian, J. S. Tay, and R. S. Pardini, “Induced oxidative stress and cell death in the A549 lung adenocarcinoma cell line by ionizing radiation is enhanced by supplementation with docosahexaenoic acid,” Nutrition and Cancer, vol. 62, no. 8, pp. 1017–1024, 2010.
[178]  K. Dittmann, C. Mayer, R. Kehlbach, M. C. Rothmund, and H. Peter Rodemann, “Radiation-induced lipid peroxidation activates src kinase and triggers nuclear EGFR transport,” Radiotherapy and Oncology, vol. 92, no. 3, pp. 379–382, 2009.
[179]  K. Dittmann, C. Mayer, B. Fehrenbacher et al., “Radiation-induced epidermal growth factor receptor nuclear import is linked to activation of DNA-dependent protein kinase,” Journal of Biological Chemistry, vol. 280, no. 35, pp. 31182–31189, 2005.
[180]  S. Girdhani, S. M. Bhosle, S. A. Thulsidas, A. Kumar, and K. P. Mishra, “Potential of radiosensitizing agents in cancer chemo-radiotherapy,” Journal of Cancer Research and Therapeutics, vol. 1, no. 3, pp. 129–131, 2005.
[181]  G. Benais-Pont, Y. M. Dupertuis, M. P. Kossovsky et al., “ω-3 Polyunsaturated fatty acids and ionizing radiation: combined cytotoxicity on human colorectal adenocarcinoma cells,” Nutrition, vol. 22, no. 9, pp. 931–939, 2006.
[182]  B. Ramanathan, K. Y. Jan, C. H. Chen, T. C. Hour, H. J. Yu, and Y. S. Pu, “Resistance to paclitaxel is proportional to cellular total antioxidant capacity,” Cancer Research, vol. 65, no. 18, pp. 8455–8460, 2005.
[183]  J. Choi, R. M. Liu, and H. J. Forman, “Adaptation to oxidative stress: quinone-mediated protection of signaling in rat lung epithelial L2 cells,” Biochemical Pharmacology, vol. 53, no. 7, pp. 987–993, 1997.
[184]  G. J. Kim, K. Chandrasekaran, and W. F. Morgan, “Mitochondrial dysfunction, persistently elevated levels of reactive oxygen species and radiation-induced genomic instability: a review,” Mutagenesis, vol. 21, no. 6, pp. 361–367, 2006.
[185]  M. A. Ogasawara and H. Zhang, “Redox regulation and Its emerging roles in stem cells and stem-like cancer cells,” Antioxidants and Redox Signaling, vol. 11, no. 5, pp. 1107–1122, 2009.
[186]  M. B. Irmak, G. Ince, M. Ozturk, and R. Cetin-Atalay, “Acquired tolerance of hepatocellular carcinoma cells to selenium deficiency: a selective survival mechanism?” Cancer Research, vol. 63, no. 20, pp. 6707–6715, 2003.
[187]  A. K. Maiti, “Gene network analysis of oxidative stress-mediated drug sensitivity in resistant ovarian carcinoma cells,” Pharmacogenomics Journal, vol. 10, no. 2, pp. 94–104, 2010.
[188]  I. Boldogh, G. Roy, M. S. Lee et al., “Reduced DNA double strand breaks in chlorambucil resistant cells are related to high DNA-PKcs activity and low oxidative stress,” Toxicology, vol. 193, no. 1-2, pp. 137–152, 2003.
[189]  B. Ramanathan, K. Y. Jan, C. H. Chen, T. C. Hour, H. J. Yu, and Y. S. Pu, “Resistance to paclitaxel is proportional to cellular total antioxidant capacity,” Cancer Research, vol. 65, no. 18, pp. 8455–8460, 2005.
[190]  T. C. Hour, C. Y. Huang, C. C. Lin et al., “Characterization of molecular events in a series of bladder urothelial carcinoma cell lines with progressive resistance to arsenic trioxide,” Anti-Cancer Drugs, vol. 15, no. 8, pp. 779–785, 2004.
[191]  K. E. de Visser and J. Jonkers, “Towards understanding the role of cancer-associated inflammation in chemoresistance,” Current Pharmaceutical Design, vol. 15, no. 16, pp. 1844–1853, 2009.
[192]  P. Borst, J. Jonkers, and S. Rottenberg, “What makes tumors multidrug resistant?” Cell Cycle, vol. 6, no. 22, pp. 2782–2787, 2007.
[193]  C. A. Schmitt, J. S. Fridman, M. Yang et al., “A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy,” Cell, vol. 109, no. 3, pp. 335–346, 2002.
[194]  G. Szakács, J. K. Paterson, J. A. Ludwig, C. Booth-Genthe, and M. M. Gottesman, “Targeting multidrug resistance in cancer,” Nature Reviews Drug Discovery, vol. 5, no. 3, pp. 219–234, 2006.
[195]  N. Voorzanger-Rousselot, L. Alberti, and J. Y. Blay, “CD40L induces multidrug resistance to apoptosis in breast carcinoma and lymphoma cells through caspase independent and dependent pathways,” BMC Cancer, vol. 6, article 75, 2006.
[196]  V. Bottero, V. Busuttil, A. Loubat et al., “Activation of nuclear factor κB through the IKK complex by the topoisomerase poisons SN38 and doxorubicin: a brake to apoptosis in HeLa human carcinoma cells,” Cancer Research, vol. 61, no. 21, pp. 7785–7791, 2001.
[197]  K. C. Das and C. W. White, “Activation of NF-κB by antineoplastic agents. Role of protein kinase C,” Journal of Biological Chemistry, vol. 272, no. 23, pp. 14914–14920, 1997.
[198]  B. Piret and J. Piette, “Topoisomerase poisons activate the transcription factor NF-κB in ACH-2 and CEM cells,” Nucleic Acids Research, vol. 24, no. 21, pp. 4242–4248, 1996.
[199]  F. R. Greten, L. Eckmann, T. F. Greten et al., “IKKβ links inflammation and tumorigenesis in a mouse model of colitis-associated cancer,” Cell, vol. 118, no. 3, pp. 285–296, 2004.
[200]  J. Dutta, Y. Fan, N. Gupta, G. Fan, and C. Gélinas, “Current insights into the regulation of programmed cell death by NF-κB,” Oncogene, vol. 25, no. 51, pp. 6800–6816, 2006.
[201]  W. Wang, J. Cassidy, V. O'Brien, K. M. Ryan, and E. Collie-Duguid, “Mechanistic and predictive profiling of 5-fluorouracil resistance in human cancer cells,” Cancer Research, vol. 64, no. 22, pp. 8167–8176, 2004.
[202]  X. Guo, B. Xu, S. Pandey et al., “Disulfiram/copper complex inhibiting NFκB activity and potentiating cytotoxic effect of gemcitabine on colon and breast cancer cell lines,” Cancer Letters, vol. 290, no. 1, pp. 104–113, 2010.
[203]  S. K. Niture and A. K. Jaiswal, “Nrf2 up-regulates anti-apoptotic protein Bcl-2 and prevents cellular apoptosis,” The Journal of Biological Chemistry, vol. 287, pp. 9873–9886, 2012.
[204]  T. C. Hour, C. Y. Huang, C. C. Lin et al., “Characterization of molecular events in a series of bladder urothelial carcinoma cell lines with progressive resistance to arsenic trioxide,” Anti-Cancer Drugs, vol. 15, no. 8, pp. 779–785, 2004.
[205]  M. Polimeni, C. Voena, J. Kopecka et al., “Modulation of doxorubicin resistance by the glucose-6-phosphate dehydrogenase activity,” Biochemical Journal, vol. 439, pp. 141–149, 2011.
[206]  H. R. Lee, J. M. Cho, D. H. Shin et al., “Adaptive response to GSH depletion and resistance to L-buthionine-(S,R)-sulfoximine: involvement of Nrf2 activation,” Molecular and Cellular Biochemistry, vol. 318, no. 1-2, pp. 23–31, 2008.
[207]  J. W. Kaspar, S. K. Niture, and A. K. Jaiswal, “Nrf2:INrf2 (Keap1) signaling in oxidative stress,” Free Radical Biology and Medicine, vol. 47, no. 9, pp. 1304–1309, 2009.
[208]  T. H. Rushmore and C. B. Pickett, “Glutathione S-transferases, structure, regulation, and therapeutic implications,” Journal of Biological Chemistry, vol. 268, no. 16, pp. 11475–11478, 1993.
[209]  A. Sau, F. Pellizzari Tregno, F. Valentino, G. Federici, and A. M. Caccuri, “Glutathione transferases and development of new principles to overcome drug resistance,” Archives of Biochemistry and Biophysics, vol. 500, no. 2, pp. 116–122, 2010.
[210]  N. Knoll, C. Ruhe, S. Veeriah et al., “Genotoxicity of 4-hydroxy-2-nonenal in human colon tumor cells is associated with cellular levels of glutathione and the modulation of glutathione S-transferase A4 expression by butyrate,” Toxicological Sciences, vol. 86, no. 1, pp. 27–35, 2005.
[211]  A. Lau, N. F. Villeneuve, Z. Sun, P. K. Wong, and D. D. Zhang, “Dual roles of Nrf2 in cancer,” Pharmacological Research, vol. 58, no. 5-6, pp. 262–270, 2008.
[212]  J. M. Cho, S. Manandhar, H. R. Lee, H. M. Park, and M. K. Kwak, “Role of the Nrf2-antioxidant system in cytotoxicity mediated by anticancer cisplatin: implication to cancer cell resistance,” Cancer Letters, vol. 260, no. 1-2, pp. 96–108, 2008.
[213]  X. J. Wang, Z. Sun, N. F. Villeneuve et al., “Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2,” Carcinogenesis, vol. 29, no. 6, pp. 1235–1243, 2008.
[214]  A. Singh, V. Misra, R. K. Thimmulappa et al., “Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer,” PLoS Medicine, vol. 3, no. 10, article e420, 2006.
[215]  P. Pettazzoni, E. Ciamporcero, C. Medana, et al., “Nuclear factor erythroid 2-related factor-2 activity controls 4-hydroxynonenal metabolism and activity in prostate cancer cells,” Free Radical Biology and Medicine, vol. 51, pp. 1610–1618, 2011.
[216]  J. T. McDonald, K. Kim, A. J. Norris et al., “Ionizing radiation activates the Nrf2 antioxidant response,” Cancer Research, vol. 70, no. 21, pp. 8886–8895, 2010.
[217]  A. Singh, M. Bodas, N. Wakabayashi, F. Bunz, and S. Biswal, “Gain of Nrf2 function in non-small-cell lung cancer cells confers radioresistance,” Antioxidants and Redox Signaling, vol. 13, no. 11, pp. 1627–1637, 2010.
[218]  S. Lee, M. J. Lim, M. H. Kim et al., “An effective strategy for increasing the radiosensitivity of Human lung Cancer cells by blocking Nrf2-dependent antioxidant responses,” Free Radical Biology and Medicine, vol. 53, no. 4, pp. 807–816, 2012.
[219]  I. B. Holland and M. A. Blight, “ABC-ATPases, adaptable energy generators fuelling transmembrane movement of a variety of molecules in organisms from bacteria to humans,” Journal of Molecular Biology, vol. 293, no. 2, pp. 381–399, 1999.
[220]  C. M. Mahaffey, H. Zhang, A. Rinna, W. Holland, P. C. Mack, and H. J. Forman, “Multidrug-resistant protein-3 gene regulation by the transcription factor Nrf2 in human bronchial epithelial and non-small-cell lung carcinoma,” Free Radical Biology and Medicine, vol. 46, no. 12, pp. 1650–1657, 2009.
[221]  C. Y. Zhang, Y. X. Feng, Y. Yu et al., “The molecular mechanism of resistance to methotrexate in mouse methotrexate-resistant cells by cancer drug resistance and metabolism superarray,” Basic and Clinical Pharmacology and Toxicology, vol. 99, no. 2, pp. 141–145, 2006.
[222]  R. Vatsyayan, P. Chaudhary, P. C. R. Lelsani et al., “Role of RLIP76 in doxorubicin resistance in lung cancer (review),” International Journal of Oncology, vol. 34, no. 6, pp. 1505–1511, 2009.
[223]  Y. C. Awasthi, R. Sharma, S. Yadav, S. Dwivedi, A. Sharma, and S. Awasthi, “The non-ABC drug transporter RLIP76 (RALBP-1) plays a major role in the mechanisms of drug resistance,” Current Drug Metabolism, vol. 8, no. 4, pp. 315–323, 2007.
[224]  K. J. Drake, J. Singhal, S. Yadav et al., “RALBP1/RLIP76 mediates multidrug resistance,” International Journal of Oncology, vol. 30, no. 1, pp. 139–144, 2007.
[225]  R. Sharma, A. Sharma, Y. Yang et al., “Functional reconstitution of Ral-binding GTPase activating protein, RLIP76, in proteoliposomes catalyzing ATP-dependent transport of glutathione conjugate of 4-hydroxynonenal,” Acta Biochimica Polonica, vol. 49, no. 3, pp. 693–701, 2002.
[226]  J. Singhal, S. S. Singhal, S. Yadav et al., “RLIP76 in defense of radiation poisoning,” International Journal of Radiation Oncology Biology Physics, vol. 72, no. 2, pp. 553–561, 2008.
[227]  T. Reya, S. J. Morrison, M. F. Clarke, and I. L. Weissman, “Stem cells, cancer, and cancer stem cells,” Nature, vol. 414, no. 6859, pp. 105–111, 2001.
[228]  C. I. Kobayashi and T. Suda, “Regulation of reactive oxygen species in stem cells and cancer stem cells,” Journal of Cellular Physiology, vol. 227, pp. 421–430, 2012.
[229]  T. M. Phillips, W. H. McBride, and F. Pajonk, “The response of CD24(-/low)/CD44t breast cancer-initiating cells to radiation,” Journal of the National Cancer Institute, vol. 98, no. 24, pp. 1777–1785, 2006.
[230]  T. Ishimoto, O. Nagano, T. Yae et al., “CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc- and thereby promotes tumor growth,” Cancer Cell, vol. 19, no. 3, pp. 387–400, 2011.
[231]  M. Diehn, R. W. Cho, N. A. Lobo et al., “Association of reactive oxygen species levels and radioresistance in cancer stem cells,” Nature, vol. 458, no. 7239, pp. 780–783, 2009.
[232]  A. Cipak, L. Mrakovcic, M. Ciz et al., “Growth suppression of human breast carcinoma stem cells by lipid peroxidation product 4-hydroxy-2-nonenal and hydroxyl radical-modified collagen,” Acta Biochimica Polonica, vol. 57, no. 2, pp. 165–171, 2010.
[233]  M. L. Guzman, R. M. Rossi, L. Karnischky et al., “The sesquiterpene lactone parthenolide induces apoptosis of human acute myelogenous leukemia stem and progenitor cells,” Blood, vol. 105, no. 11, pp. 4163–4169, 2005.
[234]  K. Ito, R. Bernardi, A. Morotti et al., “PML targeting eradicates quiescent leukaemia-initiating cells,” Nature, vol. 453, no. 7198, pp. 1072–1078, 2008.

Full-Text

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133