Skip to main content

Advertisement

Log in

Mitochondria: A novel target for the chemoprevention of cancer

  • Published:
Apoptosis Aims and scope Submit manuscript

The mitochondria have emerged as a novel target for anticancer chemotherapy. This tenet is based on the observations that several conventional and experimental chemotherapeutic agents promote the permeabilization of mitochondrial membranes in cancerous cells to initiate the release of apoptogenic mitochondrial proteins. This ability to engage mitochondrial-mediated apoptosis directly using chemotherapy may be responsible for overcoming aberrant apoptosis regulatory mechanisms commonly encountered in cancerous cells. Interestingly, several putative cancer chemopreventive agents also possess the ability to trigger apoptosis in transformed, premalignant, or malignant cells in vitro via mitochondrial membrane permeabilization. This process may occur through the regulation of Bcl-2 family members, or by the induction of the mitochondrial permeability transition. Thus, by exploiting endogenous mitochondrial-mediated apoptosis-inducing mechanisms, certain chemopreventive agents may be able to block the progression of premalignant cells to malignant cells or the dissemination of malignant cells to distant organ sites as means of modulating carcinogenesis in vivo. This review will examine cancer chemoprevention with respect to apoptosis, carcinogenesis, and the proapoptotic activity of various chemopreventive agents observed in vitro. In doing so, I will construct a paradigm supporting the notion that the mitochondria are a novel target for the chemoprevention of cancer.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Sun SY, Hail N, Jr., Lotan R. Apoptosis as a novel target for cancer chemoprevention. J Natl Cancer Inst 2004; 96: 662–672.

    PubMed  Google Scholar 

  2. Costantini P, Jocotot E, Decaudin D, Kroemer G. Mitochondrion as a novel target of anticancer chemotherapy. J Natl Cancer Inst 2000; 92: 1042–1053.

    PubMed  Google Scholar 

  3. Mignotte B, Vayssicre J-L. Mitochondria and apoptosis. Eur J Biochem 1998; 252: 1–15.

    PubMed  Google Scholar 

  4. Kroemer G, Petit P, Zamzami N, Vayssicre J-L. The biochemistry of programmed cell death. FASEB J 1995; 9: 1277–1287.

    PubMed  Google Scholar 

  5. Ferri KF, Kroemer G. Control of apoptotic DNA degradation. Nat Cell Biol 2000; 2: E63–E64.

    Article  PubMed  Google Scholar 

  6. Ferri KF, Kroemer G. Organelle-specific initiation of cell death pathways. Nat Cell Biol 2001; 3: E255–E263.

    PubMed  Google Scholar 

  7. Hengartner MO. The biochemistry of apoptosis. Nature 2000; 407: 770–776.

    PubMed  Google Scholar 

  8. Thornberry NA, Lazebnik Y. Caspases: Enemies within. Science 1998; 281: 1312–1316.

    PubMed  Google Scholar 

  9. Danial NN, Korsmeyer SJ. Cell death: Critical control points. Cell 2004; 116: 205–219.

    PubMed  Google Scholar 

  10. Kroemer G, Reed JC. Mitochondrial control of cell death. Nat Med 2000; 6: 513–519.

    PubMed  Google Scholar 

  11. Lou X, Kim CN, Yang J, Jemmerson R, Wang X. Bid, a Bcl-2 interacting protein, mediates cytochrome c release from the mitochondria in response to activation of cell surface death receptors. Cell 1998; 94: 481–490.

    PubMed  Google Scholar 

  12. Yin X-M, Wang K, Gross A, et al. Bid-deficient mice are resistant to Fas-induced hepatocellular apoptosis. Nature 1999; 400: 886–891.

    Article  PubMed  Google Scholar 

  13. Frade JM, Michaelidis TM. Origin of eukaryotic programmed cell death: A consequence of aerobic metabolism. Bioessays 1997; 19: 827–832.

    Article  PubMed  Google Scholar 

  14. Berry S. Endosymbiosis and the design of eukaryotic electron transport. Biochim Biophys Acta 2003; 1606: 57–72.

    PubMed  Google Scholar 

  15. Gray MW. Origin and evolution of mitochondrial DNA. Annu Rev Biochem 1989; 5: 25–50.

    Google Scholar 

  16. Kroemer G. Mitochondrial implication in apoptosis. Towards an endosymbiont hypothesis of apoptosis evolution. Cell Death Differ 1997; 4: 443–456.

    Article  Google Scholar 

  17. Liu X, Kim CN, Yang J, Jemmerson R, Wang X. Induction of apoptotic program in cell-free extracts: Requirement for dATP and cytochrome c. Cell 1996; 86: 147–157.

    Google Scholar 

  18. Li LY, Lou X, Wang X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature 2001; 412: 95–99.

    PubMed  Google Scholar 

  19. van Loo G, Schotte P, van Gurp M, et al. Endonuclease G: A mitochondrial protein released in apoptosis and involved in caspase-independent DNA degradation. Cell Death Differ 2001; 8: 1136–1142.

    Article  PubMed  Google Scholar 

  20. Chai J, Du C, Wu JW, Kyin S, Wang X, Shi Y. Structural and biochemical basis of apoptotic activation by Smac/DIABLO. Nature 2000; 406: 855–862.

    Article  PubMed  Google Scholar 

  21. van Loo G, van Gurp M, Depuydt B, et al. The serine protease Omi/HtrA2 is released from mitochondria during apoptosis. Omi/HtrA2 interacts with caspase-inhibitor XIAP and induces enhanced caspase activity. Cell Death Differ 2002; 9: 20–27.

    PubMed  Google Scholar 

  22. Susin SA, Lorenzo HK, Zamzami N, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999; 397: 441–446.

    PubMed  Google Scholar 

  23. Wu M, Xu L-G, Li X, Zhai Z, Shu H-B. AMID, an apoptosis-inducing factor-homologous mitochondrion-associated protein, induces caspase-independent apoptosis. J Biol Chem 2002; 277: 25617–25623.

    Article  PubMed  Google Scholar 

  24. Skulachev VP. Why are mitochondria involved in apoptosis? FEBS Lett 1996; 397: 7–10.

    Article  PubMed  Google Scholar 

  25. Skulachev VP. Mitochondrial physiology and pathology: Concepts of programmed cell death of organelles, cells, and organisms. Mol Aspects Med 1999; 20: 139–184.

    Article  PubMed  Google Scholar 

  26. Tsujinoto Y. Cell death regulation by the Bcl-2 protein family in the mitochondria. J Cell Physiol 2003; 195: 15167.

    Google Scholar 

  27. Reed JC, Jurgensmeier JM, Matsuyama S. Bcl-2 family proteins and mitochondria. Biochim Biophys Acta 1998; 1366: 127–137.

    PubMed  Google Scholar 

  28. Li H, Zhu H, Xu CJ, Yuan J. Cleavage of Bid by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 1998; 94: 491–501.

    PubMed  Google Scholar 

  29. Shimizu S, Narita M, Tsujimoto Y. Bcl-2 family proteins regulate the release of apoptogenic cytochrome c by the mitochondrial channel VDAC. Nature 1999; 399: 483–487.

    Article  PubMed  Google Scholar 

  30. Fontaine E, Bernardi P. Progress on the mitochondrial permeability transition pore: Regulation by complex I and ubiquinone analogs. J Bioenerg Biomembr 1999; 31: 335–345.

    Article  PubMed  Google Scholar 

  31. Zoratti M, Szabò I. The mitochondrial permeability transition. Biochim Biophys Acta 1995; 1241: 139–176.

    PubMed  Google Scholar 

  32. Bernardi P. Modulation of the mitochondrial cyclosporin A-sensitive permeability transition pore by the proton electrochemical gradient. Evidence that the pore can be opened by membrane depolarization. J Biol Chem 1992; 267: 8834–8839.

    PubMed  Google Scholar 

  33. Green DR, Reed JC. Mitochondria and apoptosis. Science 1998; 281: 1309–1312.

    PubMed  Google Scholar 

  34. Garrido C, Kroemer G. Life’s smile, death’s grin: Vital functions of apoptosis-executing proteins. Curr Opin Cell Biol 2004; 16: 639–646.

    Article  PubMed  Google Scholar 

  35. Pedersen PL. Mitochondrial events in the life and death of animal cells: A brief overview. J Bioenerg Biomembr 1999; 31: 291–304.

    Article  PubMed  Google Scholar 

  36. Zamzami N, Hirsch T, Dallaporta B, Petit PX, Kroemer G. Mitochondrial implication in accidental and programmed cell death: Apoptosis and necrosis. J Bioenerg Biomembr 1997; 29: 185–193.

    Article  PubMed  Google Scholar 

  37. Bertram JS. The molecular biology of cancer. Mol Aspects Med 2001; 21: 167–223.

    Article  Google Scholar 

  38. Fisher DE. Apoptosis in cancer therapy: Crossing the threshold. Cell 1994; 78: 539–542.

    Article  PubMed  Google Scholar 

  39. Martin SJ, Green DR. Apoptosis as a goal of cancer therapy. Curr Opin Oncol 1994; 6: 616–621.

    PubMed  Google Scholar 

  40. Reed JC. Mechanisms of apoptosis avoidance in cancer. Curr Opin Oncol 1999; 11: 68–75.

    Article  PubMed  Google Scholar 

  41. Adams JM, Cory S. The bcl–2 protein family: Arbiters of cell survival. Science 1998; 281: 1322–1326.

    PubMed  Google Scholar 

  42. Hong WK, Sporn MB. Recent advances in chemoprevention of cancer. Science 1997; 278: 1073–1077.

    Article  PubMed  Google Scholar 

  43. Berenblum I, Armuth V. Two independent aspects of tumor promotion. Biochim Biophys Acta 1981; 651: 51–63.

    PubMed  Google Scholar 

  44. Heidelberger C, Freeman AE, Pienta R, et al. Cell transformation by chemical agents—a review and analysis of the literature. A report of the U.S. Environmental Protection Agency Gene-Tox Program. Mutat Res 1983; 114: 283–385.

    PubMed  Google Scholar 

  45. Surh Y-J. Molecular mechanisms of chemopreventive effects of selected dietary and medicinal phenolic compounds. Mutat Res 1999; 428: 305–327.

    PubMed  Google Scholar 

  46. Guzman E, Langowski JL, Owen-Schaub L. Mad dogs, Englishmen and apoptosis: The role of cell death in UV-induced skin cancer. Apoptosis 2002; 8: 315–325.

    Article  Google Scholar 

  47. Einspahr JG, Stratton SP, Bowden GT, Alberts DS. Chemoprevention of human skin cancer. Crti Rev Oncol Hematol 2002; 41: 269–285.

    Google Scholar 

  48. Umar A, Viner JL, Hawk ET. The future of colon cancer prevention. Ann N Y Acad Sci 2001; 952: 88–108.

    PubMed  Google Scholar 

  49. Sporn MB. Approaches to prevention of epithelial cancer during the preneoplastic period. Cancer Res 1976; 36: 2699–2702.

    PubMed  Google Scholar 

  50. Kelloff GJ, Lieberman R, Steele VE, et al. Agents, biomarkers, and cohorts for chemopreventive agent development in prostate cancer. Urology 2001; 57: 46–51.

    Article  PubMed  Google Scholar 

  51. Kakizoe T. Chemoprevention of cancer—focusing on clinical trials. Jpn J Clin Oncol 2003; 33: 421–442.

    Article  PubMed  Google Scholar 

  52. Wattenberg LW. What are the critical attributes for cancer chemopreventive agents? Ann N Y Acad Sci 1995; 768: 73–81.

    PubMed  Google Scholar 

  53. Smith TJ, Hong J-Y, Wang Z-Y, Yang CS. How can carcinogenesis be inhibited? Ann N Y Acad Sci 1995; 768: 82–90.

    PubMed  Google Scholar 

  54. Kelloff GJ, Crowell JA, Steele VE, et al. Progress in cancer chemoprevention. Ann N Y Acad Sci 1999; 889: 1–13.

    PubMed  Google Scholar 

  55. Thun MJ, Henley SJ, Patrono C. Nonsteroidal anti-inflammatory drugs as anticancer agents: Mechanistic, pharmacologic, and clinical issues. J Natl Cancer Inst 2002; 20: 252–266.

    Google Scholar 

  56. Fabian CJ, Kimler BF. Beyond tamoxifen new endpoints for breast cancer chemoprevention, new drugs for breast cancer prevention. Ann N Y Acad Sci 2001; 952: 44–59.

    PubMed  Google Scholar 

  57. Modica-Napolitano JS, Steele GD, Chen LB. Aberrant mitochondria in two human colon carcinoma cell lines. Cancer Res 1989; 49: 3369–3373.

    PubMed  Google Scholar 

  58. Duchen MR. Contributions of mitochondria to animal physiology: from homeostatic sensor to calcium signalling and cell death. J Physiol 1999; 516: 1–17.

    PubMed  Google Scholar 

  59. Cantley LC, Neel BG. New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proc Natl Acad Sci USA 1999; 96: 4240–4245.

    Article  PubMed  Google Scholar 

  60. Levine AJ. p53, the cellular gatekeeper for growth and division. Cell 1997; 88: 323–331.

    PubMed  Google Scholar 

  61. Dang CV, Semenza GL. Oncogenic alterations of metabolism. Trends Biochem Sci 1999; 24: 68–72.

    Article  PubMed  Google Scholar 

  62. Cavalli LR, Liang BC. Mutagenesis, tumorigenicity, and apoptosis: Are the mitochondria involved? Mutat Res 1998; 398: 19–26.

    PubMed  Google Scholar 

  63. Spitz DR, Sim JE, Ridnour LA, Galoforo SS, Lee YJ. Glucose deprivation-induced oxidative stress in human tumor cells. A fundamental defect in metabolism? Ann N Y Acad Sci 2000; 899: 349–362.

    PubMed  Google Scholar 

  64. Sun Y, Oberley LW. Redox regulation of transcriptional activators. Free Radic Biol Med 1996; 21: 335–348.

    Article  PubMed  Google Scholar 

  65. Warburg O. On the origin of cancer cells. Science 1956; 123: 309–314.

    PubMed  Google Scholar 

  66. Hockenbery DM. A mitochondrial Achilles’ heel in cancer? Cancer Cell 2002; 2: 1–2.

    Article  PubMed  Google Scholar 

  67. Zu XL, Guppy M. Cancer metabolism: Facts, fantasy, and fiction. Biochem Biophys Res 2004; 313: 459465.

    Google Scholar 

  68. Brand K. Aerobic glycolysis by proliferating cells: Protection against oxidative stress at the expense of energy yield. J Bioenerg Biomembr 1997; 29: 355–364.

    Article  PubMed  Google Scholar 

  69. Capuano F, Guerrieri F, Papa S. Oxidative Phosphorylation enzymes in normal and neoplastic cell growth. J Bioenerg Biomembr 1997; 29: 379–384.

    Article  PubMed  Google Scholar 

  70. John AP. Dysfunctional mitochondria, not oxygen insufficiency, cause cancer cells to produce inordinate amounts of lactic acid: The impact of this on the treatment of cancer. Med Hypotheses 2001; 57: 429–431.

    Article  PubMed  Google Scholar 

  71. Krishnan B, Truong LD. Renal epithelial neoplasms: The diagnostic implications of electron microscopic study in 55 cases. Hum Pathol 2002; 33: 68–79.

    Article  PubMed  Google Scholar 

  72. Weinhouse S. Oxidative metabolism of neoplastic tissues. Cancer Res 1955; 3: 269–325.

    Google Scholar 

  73. White MT, Arya DV, Tewari KK. Biochemical properties of neoplastic cell mitochondria. J Natl Cancer Inst 1974; 53: 553–559.

    PubMed  Google Scholar 

  74. Wehrle JP, Pedersen PL. Isolation and characterization of mitochondria from malignant cells. In Cancer-Cell Organelles (Reid E, Cook GMW, Morré DJ, eds). New York: John Wiley, 1980, pp 215–225

  75. Hruban Z, Mochizuki Y, Morris HP, Slesers A. Ultrastructure of Morris renal tumors. J Natl Cancer Inst 1973; 50: 1487–1495.

    PubMed  Google Scholar 

  76. Mathupala SP, Rempel A, Pedersen PL. Glucose catabolism in cancer cells. J Biol Chem 1995; 270: 16918–16925.

    Article  PubMed  Google Scholar 

  77. Azoulay-Zohar H, Israelson A, Abu-Hamad S, Shoshan-Barmatz V. In self-defense: Hexokinase promotes voltage-dependent anion channel closure and prevents mitochondria-mediated apoptotic cell death. Biochem J 2004; 377: 347–355.

    Article  PubMed  Google Scholar 

  78. Arora KK, Parry DM, Pedersen PL. Hexokinase receptors: Preferential enzyme binding in normal cells to nonmitochondrial sites and in transformed cells to mitochondrial sites. J Bioenerg Biomembr 1992; 24: 47–53.

    Article  PubMed  Google Scholar 

  79. Crompton M. The mitochondrial permeability transition pore and its role in cell death. Biochem J 1999; 341: 233–249.

    Article  PubMed  Google Scholar 

  80. Preston TJ, Abadi A, Wilson L, Singh G. Mitochondrial contributions to cancer cell physiology: Potential for drug development. Adv Drug Del Rev 2001; 49: 45–61.

    Article  Google Scholar 

  81. Majewski N, Nogueira V, Bhaskar P, et al. Hexokinase-mitochondria interaction mediated by Akt is required to inhibit apoptosis in the presence or absence of Bax and Bak. Mol Cell 2004; 16: 819–830.

    Article  PubMed  Google Scholar 

  82. Danial NN, Gramm CF, Scorrano L, et al. BAD and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis. Nature 2003; 424: 952–956.

    Article  PubMed  Google Scholar 

  83. Guppy M, Greiner E, Brand K. The role of the Crabtree effect and an endogenous fuel in the energy metabolism of resting and proliferating thymocytes. Eur J Biochem 1993; 212: 95–99.

    Article  PubMed  Google Scholar 

  84. Sweet S, Singh G. Changes in mitochondrial mass, membrane potential, and adenosine triphosphate content during the cell cycle of human leukemic (HL-60) cells. J Cell Physiol 1999; 180: 91–96.

    Article  PubMed  Google Scholar 

  85. Dorward A, Sweet S, Moorehead R, Singh G. Mitochondrial contributions to cancer cell physiology: Redox balance, cell cycle, and drug resistance. J Bioenerg Biomembr 1997; 29: 385–392.

    Article  PubMed  Google Scholar 

  86. Chevrollier A, Loiseau D, Gauyier F, Malthièry Y, Stepien G. ANT2 expression under hypoxic conditions produces opposite cell-cycle behavior in 143B and HepG2 cancer cells. Mol Carcinog 2005; 42: 1–8.

    Article  PubMed  Google Scholar 

  87. Jiang W, Zhu Z, Bhatia N, Agarwal R, Thompson HJ. Mechanisms of energy restriction: Effects of corticosterone on cell growth, cell cycle machinery, and apoptosis. Cancer Res 2002; 62: 5280–5287.

    PubMed  Google Scholar 

  88. Zhu Z, Haegele AD, Thompson HJ. Effect of caloric restriction on pre-malignant and malignant stages of mammary carcinogenesis. Carcinogenesis 1997; 18: 1007–1012.

    Article  PubMed  Google Scholar 

  89. Wachsman JT. The beneficial effects of dietary restriction: Reduced oxidative damage and enhanced apoptosis. Mutat Res 1996; 350: 25–34.

    PubMed  Google Scholar 

  90. Brit DF, Pelling JC, White LT, Dimitroff K, Barnett T. Influence of diet and calorie restriction on the initiation and promotion of skin carcinogenesis in the SENCAR mouse model. Cancer Res 1991; 51: 1851–1854.

    PubMed  Google Scholar 

  91. Kolaja KL, Bunting KA, Klaunig JE. Inhibition of tumor promotion and hepatocellular growth by dietary restriction in mice. Carcinogenesis 1996; 17: 1657–1664.

    PubMed  Google Scholar 

  92. Kritchevsky D, Klurfeld DM. Interaction of fiber and energy registration in experimental colon carcinogens. Cancer Lett 1997; 114: 51–52.

    Article  PubMed  Google Scholar 

  93. Thompson HJ, Zhu Z, Jiang W. Identification of the apoptosis activation cascade induced in mammary carcinomas by energy restriction. Cancer Res 2004; 64: 1541–1545.

    PubMed  Google Scholar 

  94. Tirosh O, Aronis A, Zusman I, et al. Mitochondrion-mediated apoptosis is enhanced in long-lived aMUPA transgenic mice and calorically restricted wild-type mice. Exp Gerontol 2003; 38: 955–963.

    Article  PubMed  Google Scholar 

  95. Birch-Machin MA. Mitochondria and skin disease. Exp Dermatol 2000; 25: 141–146.

    Article  Google Scholar 

  96. Penta JS, Johnson FM, Wachsman JT, Copeland WC. Mitochondrial DNA in human malignancy. Mutat Res 2001; 488: 119–133.

    Article  PubMed  Google Scholar 

  97. Attardi G, Schatz G. Biogenesis of mitochondria. Annu Rev Cell Biol 1988; 4: 289–333.

    Article  PubMed  Google Scholar 

  98. Wallace DC. Mitochondrial diseases in man and mouse. Science 1999; 283: 1482–1488.

    Article  PubMed  Google Scholar 

  99. Carew JS, Huang P. Mitochondrial defects in cancer. Mol Cancer 2002; 9: 1–12.

    Google Scholar 

  100. Wei Y-H, Kao S-H, Lee H-C. Simultaneous increase of mitochondrial DNA deletions and lipid peroxidation in human aging. Ann N Y Acad Sci 1996; 786: 24–42.

    PubMed  Google Scholar 

  101. Yakes MF, Van Houten B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc Natl Acad Sci USA 1997; 94: 514–519.

    Article  PubMed  Google Scholar 

  102. Bianchi NO, Bianchi MS, Richard SM. Mitochondrial genome instability in human cancers. Mutat Res 2001; 448: 9–23.

    Google Scholar 

  103. Carew JS, Zhou Y, Albitar M, Carew JD, Keating MJ, Huang P. Mitochondrial DNA mutations in primary leukemia cells after chemotherapy: Clinical significance and therapeutic implications. Leukemia 2003; 17: 1437–1447.

    Article  PubMed  Google Scholar 

  104. Harris AL. Hypoxia-a key regulatory factor in tumour growth. Nat Rev Cancer 2002; 2: 38–47.

    Article  PubMed  Google Scholar 

  105. Vaupel P, Thews O, Kellether DK, Hoeckel M. Current status of knowledge of critical issues in tumor oxygenation-results from 25 years of research in tumor pathophysiology. Adv Exp Med Biol 1998; 454: 591–602.

    PubMed  Google Scholar 

  106. Brown GC. Control of respiration and ATP synthesis in mammalian mitochondria and cells. Biochem J 1992; 284: 1–13.

    PubMed  Google Scholar 

  107. Hatefi Y. The mitochondrial electron transport and oxidative phosphorylation system. Annu Rev Biochem 1985; 54: 1015–1069.

    Article  PubMed  Google Scholar 

  108. St-Pierre J, Brand MD, Boutilier RG. Mitochondria as ATP consumers: Cellular treason in anoxia. Proc Natl Acad Sci USA 2000; 97: 8670–8674.

    Article  PubMed  Google Scholar 

  109. Buchet K, Godinot C. Functional F1-ATPase essential in maintaining growth and membrane potential of human mitochondrial DNA-depleted ρ0 cells. J Biol Chem 1998; 273: 22983–22989.

    Article  PubMed  Google Scholar 

  110. Giraud S, Bonod-Bidaud C, Wesolowski-Louvel M, Stepien G. Expression of human ANT2 gene in highly proliferative cells: GRBOX, a new transcriptional element, is involved in the regulation of glycolytic ATP import into mitochondria. Journal of Molecular Biology 1998; 281: 409-418.

    Article  PubMed  Google Scholar 

  111. Anderson KM, Harris JE. Is induction of type 2 programmed cell death in cancer cells from solid tumors directly related to mitochondrial mass? Med Hypotheses 2001; 57: 87–90.

    Article  PubMed  Google Scholar 

  112. Grad JM, Cepero E, Boise LH. Mitochondria as targets for established and novel anti-cancer agents. Drug Resist Updates 2001; 4: 85–91.

    Article  Google Scholar 

  113. Singh KK, Russell J, Sigala B, Zhang Y, Wiliams J, Keshav KF. Mitochondrial DNA determines the cellular response to cancer therapeutic agents. Oncogene 1999; 18: 6641–6646.

    Article  PubMed  Google Scholar 

  114. Hail N, Jr., Youssef EM, Lotan R. Evidence supporting a role for mitochondrial respiration in apoptosis induction by the synthetic retinoid CD437. Cancer Res 2001; 61: 6698–6702.

    PubMed  Google Scholar 

  115. Hail N, Jr., Lotan R. Mitochondrial respiration is uniquely associated with the prooxidant and apoptotic effects of N-(4-hydroxyphenyl)retinamide. J Biol Chem 2001; 276: 45614–45621.

    Article  PubMed  Google Scholar 

  116. Hail N, Jr., Lotan R. Examining the role of mitochondrial respiration in vanilloid-induced apoptosis. J Natl Cancer Inst 2002; 94: 1281–1292.

    PubMed  Google Scholar 

  117. Hail N, Jr., Lotan R. Apoptosis induction by the natural product cancer chemopreventive agent deguelin is mediated through the inhibition of mitochondrial respiration. Apoptosis 2004; 9: 437–447.

    Article  PubMed  Google Scholar 

  118. Li N, Ragheb K, Lawler G, et al. Mitochondrial complex I inhibitor rotenone induces apoptosis through enhancing mitochondrial reactive species production. J Biol Chem 2003; 278: 8516–8525.

    Article  PubMed  Google Scholar 

  119. Weber T, Dalen H, Andera L, et al. Mitochondria play a central role in apoptosis induced by α-tocopheryl succinate, an agent with antineoplastic activity: Comparison with receptor-mediated pro-apoptotic signaling. Biochemistry 2003; 42: 4277–4291.

    Article  PubMed  Google Scholar 

  120. Wang XF, Witting PK, Salvatore BA, Neuzil J. Vitamin E analogs trigger apoptosis in HER2/erbB2-overexpressing breast cancer cells by signaling via the mitochondrial pathway. Biochem Biophys Res 2005; 326: 282–289.

    Article  Google Scholar 

  121. Joshi B, Li L, Taffe BG, et al. Apoptosis induction by a novel anti-prostate cancer compound, BMD188 (a fatty acid containing hydroxamic acid), requires the mitochondrial respiratory chain. Cancer Res 1999; 59: 4343–4355.

    PubMed  Google Scholar 

  122. Poppe M, Reimertz C, Munstermann G, Kogel D, Prehn JH. Ceramide-induced apoptosis of D283 medulloblastoma cells requires mitochondrial respiratory chain activity but occurs independently of caspases and is not sensitive to Bcl-xL overexpression. J Neurochem 2002; 82: 482–494.

    Article  PubMed  Google Scholar 

  123. Higuchi M, Aggarwal BB, Yeh ET. Activation of CPP32-like protease in tumor necrosis factor-induced apoptosis is dependent on mitochondrial function. J Clin Invest 1997; 99: 1751–1758.

    PubMed  Google Scholar 

  124. Kim J-Y, Kim Y-H, Chang I, et al. Resistance of mitochondrial DNA-deficient cells to TRAIL: Role of Bax in TRAIL-induced apoptosis. Oncogene 2002; 21: 3139–3148.

    Article  PubMed  Google Scholar 

  125. Cardoso CM, Moreno AJ, Almeida LM, Custodio JB. Comparison of the changes in adenine nucleotides of rat liver mitochondria induced by tamoxifen and 4-hydroxytamoxifen. Toxicol in vitro 2003; 17: 663–670.

    Article  PubMed  Google Scholar 

  126. Fang N, Casida JE. Anticancer action of cubè insecticide: Correlation for rotenoid constituents between inhibition of NADH: Ubiquinone oxidoreductase and induced ornithine decarboxylase activities. Proc Natl Acad Sci USA 1998; 95: 3380–3384.

    Article  PubMed  Google Scholar 

  127. Chudapongse P, Janthasoot W. Mechanism of inhibitory action of capsaicin on energy metabolism by rat liver mitochondria. Biochem Pharmacol 1981; 30: 735–740.

    Article  PubMed  Google Scholar 

  128. Degli Esposti M, Crimi M, Ghelli A. Natural variation in the potency and binding sites of mitochondrial quinone-like inhibitors. Biochem Soc Trans 1994; 22: 209–213.

    PubMed  Google Scholar 

  129. Okun JG, Lümmen P, Brandt U. Three classes of inhibitors share a common binding domain in mitochondrial complex I (NADH:Ubiquinone oxidoreductase). J Biol Chem 1999; 274: 2625–2630.

    Article  PubMed  Google Scholar 

  130. Armstrong JS, Hornung B, Lecane P, Jones DP, Knox SJ. Rotenone-induced G2/M cell cycle arrest and apoptosis in a human B lymphoma cell line PW. Biochem Biophys Res 2001; 289: 973–978.

    Article  Google Scholar 

  131. Mills KI, Woodgate LJ, Gilkes AF, et al. Inhibition of mitochondrial function in HL60 cells is associated with an increased apoptosis and expression of CD14. Biochem Biophys Res 1999; 263: 294–300.

    Article  Google Scholar 

  132. Kitamura Y, Inden M, Miyamara A, Kakimura J, Tanigucgi T, Shimohama S. Possible involvement of both mitochondria- and endoplasmic reticulum-dependent caspase pathways in rotenone-induced apoptosis in human neuroblastoma SH-SY5Y cells. Neurosci Lett 2002; 333: 25–28.

    Article  PubMed  Google Scholar 

  133. Gerhäuser C, Lee SK, Kosmeder JW, et al. Regulation of ornithine decarboxylase induction by deguelin, a natural product cancer chemopreventive agent. Cancer Res 1997; 57: 3429–3435.

    PubMed  Google Scholar 

  134. Hail N, Jr. Mechanisms of vanilloid-induced apoptosis. Apoptosis 2003; 8: 251–262.

    Article  PubMed  Google Scholar 

  135. Pelicano H, Feng L, Zhou Y, et al. Inhibition of mitochondrial respiration. A novel strategy to enhance drug-induced apoptosis in human leukemia cells by a reactive oxygen species-mediated mechanism. J Biol Chem 2003; 278: 37832–37839.

    Article  PubMed  Google Scholar 

  136. Delia D, Aiello A, Lombardi L, et al. N-(4-Hydroxyphenyl)retinamide induces apoptosis of malignant hemopoietic cell lines including those unresponsive to retinoic acid. Cancer Res 1993; 53: 6036–6041.

    PubMed  Google Scholar 

  137. Hail N, Jr., Lotan R. Mitochondrial permeability transition is a central coordinating event in N-(4-hydroxyphenly)retinamide-induced apoptosis. Cancer Epidemiol Biomarkers Prev 2000; 9: 1293–1301.

    PubMed  Google Scholar 

  138. Oridate N, Lotan D, Xu XC, Hong WK, Lotan R. Differential induction of apoptosis by all-trans-retinoic acid and N-(4-hydroxyphenyl)retinamide in human head and neck squamous cell carcinoma cell lines. Clin Cancer Res 1996; 2: 855–863.

    PubMed  Google Scholar 

  139. Maurer B, Metelitsa L, Seeger R, Cabot M, Reynolds C. Increased of ceramide and induction of mixed apoptosis/necrosis by N-(4-hydroxyphenyl)retinamide in neuroblastoma cell lines. J Natl Cancer Inst 1999; 91: 1138–1146.

    PubMed  Google Scholar 

  140. Sun S-Y, Yue P, Lotan R. Induction of apoptosis by N-(4-hydroxyphenyl)retinamide and its association with reactive oxygen species, nuclear retinoic acid receptors, and apoptosis related genes in human prostate carcinoma cells. Mol Pharmacol 1999; 55: 403–410.

    PubMed  Google Scholar 

  141. Huang Y, He Q, Hillman MJ, Rong R, Sheikh MS. Sulindac sulfide-induced apoptosis involves death receptor 5 and the caspase 8-dependent pathway in human colon and prostate cancer cells. Cancer Res 2001; 61: 6918–6924.

    PubMed  Google Scholar 

  142. Lim JT, Piazza GA, Han EK, et al. Sulindac derivatives inhibit growth and induce apoptosis in human prostate cancer cell lines. Biochem Pharmacol 1999; 58: 1097–1107.

    Article  PubMed  Google Scholar 

  143. Rahman MA, Dhar DK, Masunaga R, Yamanoi A, Kohno H, Nagasue N. Sulindac and exisulind exhibit a significant antiproliferative effect and induce apoptosis in human hepatocellular carcinoma cell lines. Cancer Res 2000; 60: 2085–2089.

    PubMed  Google Scholar 

  144. Zhang L, Yu J, Park BH, Kinzler KW, Vogelstein B. Role of BAX in the apoptotic response to anticancer agents. Science 2000; 290: 989–992.

    PubMed  Google Scholar 

  145. Hsu AL, Ching TT, Wang DS. The cyclooxygenase-2 inhibitor celecoxib induces apoptosis by blocking Akt activation in human prostate cancer cells independently of Bcl-2. J Biol Chem 2000; 275: 11397–11403.

    Article  PubMed  Google Scholar 

  146. Dietze EC, Caldwell LE, Grupin SL, Mancini M, Seewaldt VL. Tamoxifen but not 4-hydroxytamoxifen initiates apoptosis in p53(-) normal human mammary epithelial cells by inducing mitochondrial depolarization. J Biol Chem 2001; 276: 5384–5394.

    Article  PubMed  Google Scholar 

  147. Ferlini C, Scambia G, Marone M, et al. Tamoxifen induces oxidative stress and apoptosis in oestrogen receptor-negative human cancer cell lines. Br J Cancer 1999; 79: 257–263.

    PubMed  Google Scholar 

  148. Kang Y, Cortina R, Perry RR. Role of c-myc in tamoxifen-induced apoptosis estrogen-independent breast cancer cells. J Natl Cancer Inst 1996; 88: 279–284.

    PubMed  Google Scholar 

  149. Delia D, Aiello A, Meroni L, Nicolini M, Reed JC, Pierotti MA. Role of antioxidants and intracellular free radicals in retinamide-induced cell death. Carcinogenesis 1997; 18: 943–948.

    Article  PubMed  Google Scholar 

  150. Oridate N, Suzuki S, Higuchi M, Mitchell MF, Hong WK, Lotan R. Involvement of reactive oxygen species in N-(4-hydroxyphenyl)retinamide-induced apoptosis in cervical carcinoma cells. J Natl Cancer Inst 1997; 89: 1191–1198.

    Article  PubMed  Google Scholar 

  151. Suzuki S, Higuchi M, Proske RJ, Oridate N, Hong WK, Lotan R. Implication of mitochondria-derived reactive oxygen species, cytochrome c and caspase-3 in N-(4-hydroxyphenyl)retinamide-induced apoptosis in cervical carcinoma cells. Oncogene 1999; 18: 6380–6387.

    Article  PubMed  Google Scholar 

  152. Chan TA, Morin P, Vogelstein B, Kinzler KW. Mechanisms underlying nonsteroidal antiinflammatory drug-mediated apoptosis. Proc Natl Acad Sci U S A 1998; 95: 681–686.

    Article  PubMed  Google Scholar 

  153. Kusuhara H, Komatsu H, Sumichika H, Sugahara K. Reactive oxygen species are involved in the apoptosis induced by nonsteroidal anti-inflammatory drugs in cultured gastric cells. Eur J Pharmacol 1999; 383: 331–337.

    Article  PubMed  Google Scholar 

  154. Yang GY, Liao J, Li C, et al. Effect of black and green tea polyphenols on c-jun phosphorylation and H2O2 production in transformed and non-transformed human bronchial cell lines: Possible mechanisms of cell growth inhibition and apoptosis induction. Carcinogenesis 2000; 21: 2035–2039.

    Article  PubMed  Google Scholar 

  155. Morin D, Barthelemy S, Zini R, Labidalle S, Tillement JP. Curcumin induces the mitochondrial permeability transition pore mediated by membrane protein thiol oxidation. FEBS Lett 2001; 495: 131–136.

    Article  PubMed  Google Scholar 

  156. Ikeda T, Sporn MB, Honda T, Gribble GW, Kufe D. The novel triterpenoid CDDO and its derivatives induces apoptosis by disruption of intracellular redox balance. Cancer Res 2003; 63: 5551–5558.

    PubMed  Google Scholar 

  157. Hail N, Jr., Konopleva M, Sporn M, Lotan R, Andreeff M. Evidence supporting a role for calcium in apoptosis induction by the synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO). J Biol Chem 2004; 279: 11179–11187.

    Article  PubMed  Google Scholar 

  158. Mandlekar S, Yu R, Tan TH, Kong AN. Activation of caspase-3 and c-Jun NH2-terminal kinase-1 signaling pathways in tamoxifen-induced apoptosis of human breast cancer cells. Cancer Res 2000; 60: 5995–6000.

    PubMed  Google Scholar 

  159. Macho A, Calcado MA, Muñoz-Blanco J, et al. Selective induction of apoptosis by capsaicin in transformed cells: The role of reactive oxygen species and calcium. Cell Death Differ 1999; 6: 155–165.

    Article  PubMed  Google Scholar 

  160. Macho A, Blázquez M-V, Navas P, Muñoz E. Induction of apoptosis by vanilloid compounds does not require de novo gene transcription and activator protein 1 activity. Cell Growth Differ 1998; 9: 277–286.

    PubMed  Google Scholar 

  161. Richeux F, Cascante M, Ennamany R, et al. Implication of oxidative stress and inflammatory process in the toxicity of capsaicin in human endothelial cells: Lack of DNA strand breakage. Toxicology 2000; 147: 41–49.

    Article  PubMed  Google Scholar 

  162. Macho A, Lucena C, Calzado MA, et al. Phorboid 20-homovanillates induce apoptosis through a VR1-independent mechanism. Chem Biol 2000; 7: 483–492.

    Article  PubMed  Google Scholar 

  163. Isenberg JS, Klaunig JE. Role of the mitochondrial membrane permeability transition (MPT) in rotenone-induced apoptosis in liver cells. Toxicol Sci 2000; 53: 340–351.

    Article  PubMed  Google Scholar 

  164. Kang YH, Lee E, Choi MK, et al. Role of reactive oxygen species in the induction of apoptosis by alpha-tocopheryl succinate. Int J Cancer 2004; 112: 385–392.

    Article  PubMed  Google Scholar 

  165. Kogure K, Morita M, Nakashima S, Hama S, Tokumura A, Fukuzawa K. Super-oxide is responsible for apoptosis in rat vascular smooth muscle cells induced by α-tocopheryl hemisuccinate. Biochim Biophys Acta 2001; 1528: 25–30.

    PubMed  Google Scholar 

  166. Andoh T, Lee SY, Chiueh CC. Preconditioning regulation of bcl-2 and p66shc by human NOS1 enhances tolerance to oxidative stress. FASEB J 2000; 14: 2144–2146.

    PubMed  Google Scholar 

  167. Aoki M, Nata T, Morishita R, et al. Endothelial apoptosis induced by oxidative stress through activation of NF-kB: Antiapoptotic effect of antioxidant agents on endothelial cells. Hypertension 2001; 38: 48–55.

    PubMed  Google Scholar 

  168. Bojes HK, Datta K, Xu J, et al. Bcl-xL overexpression attenuates glutathione depletion in FL5.12 cells following interleukin-3 withdrawal. Biochem J 1997; 325: 315–319.

    PubMed  Google Scholar 

  169. Crawford MJ, Krishnamoorthy RR, Rudick VL, et al. Bcl-2 overexpression protects photooxidative stress-induced apoptosis of photoreceptor cells via NF-kB preservation. Biochem Biophys Res 2001; 281: 1304–1312.

    Article  Google Scholar 

  170. Sonoda Y, Watanabe S, Matsumoto Y, Aizu-Yokota E, Kasahara T. FAK is the upstream signal protein of the phosphatidylinositol 3-kinase-Akt survival pathway in hydrogen peroxide-induced apoptosis of a human glioblastoma cell line. J Biol Chem 1999; 274: 10566–10570.

    Article  PubMed  Google Scholar 

  171. Longoni B, Boschi E, Demontis GC, Ratto GM, Mosca F. Apoptosis and adaptive responses to oxidative stress in human endothelial cells exposed to cyclosporin A correlate with BCL-2 expression levels. FASEB J 2001; 15: 731–740.

    Article  PubMed  Google Scholar 

  172. Zhang W, Couldwell WT, Song H, Takano T, Lin JH, Nedergaard M. Tamoxifen-induced enhancement of calcium signaling in glioma and MCF-7 breast cancer cells. Cancer Res 2000; 60: 5395–5400.

    PubMed  Google Scholar 

  173. Zhou XM, Wong BC, Fan XM, et al. Non-steroidal anti-inflammatory drugs induce apoptosis in gastric cancer cells through up-regulation of bax and bak. Carcinogenesis 2001; 22: 1393–1397.

    Article  PubMed  Google Scholar 

  174. Li Y, Upadhyay S, Bhuiyan M, Sarkar FH. Induction of apoptosis in breast cancer cells MDA-MB-231 by genistein. Oncogene 1999; 18: 3166–3172.

    Article  PubMed  Google Scholar 

  175. Jung M-Y, Kang H-J, Moon A. Capsaicin-induced apoptosis in SK-Hep-1 hepatocarcinoma cells involves Bcl-2 downregulation and caspase-3 activation. Cancer Lett 2001; 165: 139–145.

    Article  PubMed  Google Scholar 

  176. Chun K-H, Kosmeder JW, Sun S, et al. Effects of deguelin on the phosphatidylinositol 3-kinase/Akt pathway and apoptosis in premalignant human bronchial epithelial cells. J Natl Cancer Inst 2003; 95: 291–302.

    PubMed  Google Scholar 

  177. Davis JN, Kucuk O, Sarkar FH. Genistein inhibits NF-kB activation in prostate cancer cells. Nutr Cancer 1999; 35: 167–174.

    Article  PubMed  Google Scholar 

  178. Ahmad N, Gupta S, Mukhtar H. Green tea polyphenol epigallocatechin-3-gallate differentially modulates nuclear factor kappa B in cancer cells versus normal cells. Arch Biochem Biophys 2000; 376: 338–346.

    Article  PubMed  Google Scholar 

  179. Holmes-McNary M, Baldwin AS, Jr. Chemopreventive properties of trans-resveratrol are associated with inhibition of activation of the IkappaB kinase. Cancer Res 2000; 60: 3477–3483.

    PubMed  Google Scholar 

  180. Farina AR, Masciulli MP, Tacconelli A, et al. All-trans-retinoic acid induces nuclear factor kappaB activation and matrix metalloproteinase-9 expression and enhances basement membrane invasivity of differentiation-resistant human SK-N-BE 9N neuroblastoma cells. Cell Growth Differ 2002; 13: 343–354.

    PubMed  Google Scholar 

  181. Nawata H, Maeda Y, Sumimoto Y, Miyatake J, Kanamaru A. A mechanism of apoptosis induced by all-trans retinoic acid on adult T-cell leukemia cells: A possible involvement of the Tax/NF-kappaB signaling pathway. Leuk Res 2001; 25: 323–331.

    Article  PubMed  Google Scholar 

  182. Han SS, Keum Y-S, Seo H-J, Chun K-S, Lee SS, Surh Y-J. Capsaicin suppresses phorbol ester-induced activation of NF-kB/Rel and AP-1 transcription factors in mouse epidermis. Cancer Lett 2001; 164: 119–126.

    Article  PubMed  Google Scholar 

  183. Cipriani B, Borsellino G, Knowles H, et al. Curcumin inhibits activation of Vg9Vd2 T cells by phosphoantigens and induces apoptosis involving apoptosis-inducing factor and large scale DNA fragmentation. J Immunol 2001; 167: 3454–3462.

    PubMed  Google Scholar 

  184. Neuzil J, Schröder A, von Hundelshausen P, et al. Inhibition of inflammatory endothelial responses by a pathway involving caspase activation and p65 cleavage. Biochemistry 2001; 40: 4686–4692.

    Article  PubMed  Google Scholar 

  185. Dalen H, Neuzil J. α-Tocopheryl succinate sensitizes T lymphoma cells to TRAIL killing by suppressing NF-kB activation. Br J Cancer 2003; 88: 153–158.

    Article  PubMed  Google Scholar 

  186. Van Antwerp DJ. Suppression of TNF-α-induced apoptosis by NF-kB. Science 1997; 274: 787–789.

    Article  Google Scholar 

  187. Vollgraf U, Wegner M, Richter-Landsberg C. Activation of AP-1 and nuclear factor-kappa B transcription factors is involved in hydrogen peroxide-induced apoptotic cell death of oligodendrocytes. J Neurochem1999; 73: 2501–2509.

    Article  PubMed  Google Scholar 

  188. Fleury C, Mignotte B, Vayssière J-L. Mitochondrial reactive species in cell death signaling. Biochimie 2002; 84: 131–141.

    Article  PubMed  Google Scholar 

  189. Turrens JF. Superoxide production by the mitochondrial respiratory chain. Biosci Rep 1997; 17: 3–7.

    Article  PubMed  Google Scholar 

  190. Chance B, Sies H, Boveris A. Hydroperoxide metabolism in mammalian organs. Physiol Rev 1979; 59: 527–605.

    PubMed  Google Scholar 

  191. Skulachev VP. Membrane-linked systems preventing superoxide formation. Biosci Rep 1997; 17: 347–365.

    Article  PubMed  Google Scholar 

  192. Kovacic P, Jacintho DJ. Mechanisms of carcinogenesis: Focus on oxidative stress and electron transfer. Curr Med Chem2001; 8: 73–796.

    Google Scholar 

  193. Galati G, Sabzevari O, Wilson JX, O’Brien PJ. Prooxidant activity and cellular effects of the phenoxyl radicals of dietary flavonoids and other polyphenolics. Toxicology 2002; 177: 91–104.

    Article  PubMed  Google Scholar 

  194. Galati G, O’Brien PJ. Potential toxicity of flavonoids and other dietary phenolics: Significance for their chemopreventive and anticancer properties. Free Radic Biol Med 2004; 37: 287–303.

    Article  PubMed  Google Scholar 

  195. Kagan VE, Tyurina YY. Recycling and redox cycling of phenolic antioxidants. Ann N Y Acad Sci 1998; 854: 425–434.

    PubMed  Google Scholar 

  196. Kinnula VL, Crapo JD. Superoxide dismutases in malignant cells and human tumors. Free Radic Biol Med 2002; 36: 718–744.

    Article  Google Scholar 

  197. Wallace KB, Starkov AA. Mitochondrial targets of drug toxicity. Annu Rev Pharmacol Toxicol 2000; 40: 353–388.

    Article  PubMed  Google Scholar 

  198. Kagan VE, Serbinova EA, Packer L. Generation and recycling of radicals from phenolic antioxidants. Arch Biochem Biophys 1990; 280: 33–39.

    Article  PubMed  Google Scholar 

  199. Galati G, Teng S, Moridani MY, Chan TS, O’Brien PJ. Cancer chemoprevention and apoptosis mechanisms induced by dietary polyphenolics. Drug Metab Drug Interact 2000; 17: 311–349.

    Google Scholar 

  200. Degli Esposti M. Inhibitors of NADH-ubiquinone reductase: An overview. Biochim Biophys Acta 1998; 1364: 222–235.

    PubMed  Google Scholar 

  201. Hodnick WF, Duval DL, Pardini RS. Inhibition of mitochondrial respiration and cyanide-stimulated generation of reactive oxygen species by selected flavonoids. Biochem Pharmacol 1994; 47: 573–580.

    Article  PubMed  Google Scholar 

  202. Miyoshi H. Structure-activity relationships of some complex I inhibitors. Biochim Biophys Acta 1998; 1364: 236–244.

    PubMed  Google Scholar 

  203. Satoh T, Miyoshi H, Sakamoto K, Iwamura H. Comparison of the inhibitory action of the synthetic capsaicin analogues with various NADH-ubiquinone oxidoreductases. Biochim Biophys Acta 1996; 1273: 21–30.

    PubMed  Google Scholar 

  204. Shimomura Y, Kawada T, Suzuki M. Capsaicin and its analogs inhibit the activity of NADH-coenzyme Q oxidoreductase of the mitochondrial respiratory chain. Arch Biochem Biophys 1989; 270: 573–577.

    Article  PubMed  Google Scholar 

  205. Yagi T. Inhibition by capsaicin of NADH-quinone oxidoreductase is correlated with the presence of energy-coupling site 1 in various organisms. Arch Biochem Biophys 1990; 281: 305–311.

    Article  PubMed  Google Scholar 

  206. Skulachev VP. Role of uncoupled and non-coupled oxidations in maintenance of safely low levels of oxygen and its one-electron reductants. Q Rev Biophys 1996; 29: 169–202.

    PubMed  Google Scholar 

  207. Rose P, Armstrong JS, Chua YL, Ong CN, Whiteman M. β-Phenylethyl isothiocyanate mediated apoptosis; contribution of Bax and the mitochondrial death pathway. Int J Biochem Cell Biol 2005; 37: 100–119.

    Article  PubMed  Google Scholar 

  208. Nakamura Y, Kawakami M, Yoshihiro A, et al. Involvement of the mitochondrial death pathway in chemopreventive benzyl isothiocyanate-induced apoptosis. J Biol Chem 2002; 277: 8492–8499.

    Article  PubMed  Google Scholar 

  209. Cardoso CM, Custodio JB, Almeida LM, Moreno AJ. Mechanisms of the deleterious effects of tamoxifen on mitochondrial respiration rate and phosphorylation efficiency. Toxicology and Applied Pharmacology 2001; 176: 145–152.

    Article  PubMed  Google Scholar 

  210. Fang N, Casida JE. Cubè resin insecticide: Identification and biological activity of 29 rotenoid constituents. J Agric Food Chem 1999; 47: 2130–2136.

    Article  PubMed  Google Scholar 

  211. Rowlands JC, Casida JE. NADH:Ubiquinone oxidoreductase inhibitors block induction of ornithine decarboxylase activity in MCF-7 human breast cancer cells. Pharmacol Toxicol 1998; 83: 214–219.

    PubMed  Google Scholar 

  212. Marchetti P, Zamzami N, Joseph B, et al. The novel retinoid 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphtalene carboxylic acid can trigger apoptosis through a mitochondrial pathway independent of the nucleus. Cancer Res 1999; 59: 6257–6266.

    PubMed  Google Scholar 

  213. Ligeret H, Barthelemy S, Zini R, Tillement JP, Labidalle S, Morin D. Effects of curcumin and curcumin derivatives on mitochondrial permeability transition pore. Free Radic Biol Med 2004; 36: 919–929.

    Article  PubMed  Google Scholar 

  214. Belzacq AS, El Hamel C, Vieira HL, et al. Adenine nucleotide translocator mediates the mitochondrial membrane permeabilization induced by lonidamine, arsenite and CD437. Oncogene 2001; 20: 7579–7587.

    Article  PubMed  Google Scholar 

  215. Boya P, Morales MC, Gonzalez-Polo RA, et al. The chemopreventive agent N-(4-hydroxyphenyl)retinamide induces apoptosis through a mitochondrial pathway regulated by proteins from the Bcl-2 family. Oncogene 2003; 22: 6220-6230.

    Article  PubMed  Google Scholar 

  216. Hayon T, Dvilansky A, Oriev L, Nathan I. Non-steroidal antiestrogens induce apoptosis in HL60 and MOLT3 leukemic cells; involvement of reactive oxygen radicals and protein kinase C. Anticancer Res 1999; 19: 2089–2093.

    PubMed  Google Scholar 

  217. Zhang GJ, Kimijima I, Onda M, et al. Tamoxifen-induced apoptosis in breast cancer cells relates to down-regulation of bcl-2, but not bax and bcl-XL, without alteration of p53 protein levels. Clin Cancer Res 1999; 5: 2971–2977.

    PubMed  Google Scholar 

  218. Heerdt BG, Houston MA, Anthony GM, Augenlicht LH. Initiation of growth arrest and apoptosis of MCF-7 mammary carcinoma cells by tributyrin, a triglyceride analogue of the short-chain fatty acid butyrate, is associated with mitochondrial activity. Cancer Res 1999; 59: 1584–1591.

    PubMed  Google Scholar 

  219. Yoon HS, Moon SC, Kim ND, Park BS, Jeong MH, Yoo YH. Genistein induces apoptosis of RPE-J cells by opening mitochondrial PTP. Biochem Biophys Res 2000; 276: 151–156.

    Article  Google Scholar 

  220. Hu R, Kim BR, Chen C, Hebbar V, Kong AN. The roles of JNK and apoptotic signaling pathways in PEITC-mediated responses in human HT-29 colon adenocarcinoma cells. Carcinogenesis 2003; 24: 1361–1367.

    Article  PubMed  Google Scholar 

  221. Zimmermann KC, Waterhouse NJ, Goldstein JC, Schuler M, Green DR. Aspirin induces apoptosis through release of cytochrome c from mitochondria. Neoplasia 2000; 2: 505–513.

    Article  PubMed  Google Scholar 

  222. Pique M, Barragan M, Dalmau M, Bellosillo B, Pons G, Gil J. Aspirin induces apoptosis through mitochondrial cytochrome c release. FEBS Lett 2000; 480: 193–196.

    Article  PubMed  Google Scholar 

  223. Zhang Z, Lai GH, Sirica AE. Celecoxib-induced apoptosis in rat cholangiocarcinoma cells mediated by Akt inactivation and Bax translocation. Hepatology 2004; 39: 1028–1037.

    Article  PubMed  Google Scholar 

  224. Jendrossek V, Handrick R, Belka C. Celecoxib activates a novel mitochondrial apoptosis signaling pathway. FASEB J 2003; 17: 1547–1549.

    PubMed  Google Scholar 

  225. Aggarwal S, Taneja N, Lin L, Orringer MB, Rehemtulla A, Beer DG. Indomethacin-induced apoptosis in esophageal adenocarcinoma cells involves upregulation of Bax and translocation of mitochondrial cytochrome C independent of COX-2 expression. Neoplasia 2000; 2: 346–356.

    Article  PubMed  Google Scholar 

  226. Waddell WR. Stimulation of apoptosis by sulindac and piroxicam. Clin Sci 1998; 95: 385–388.

    Article  PubMed  Google Scholar 

  227. Chen C, Shen G, Hebbar V, Hu R, Owuor ED, Kong AN. Epigallocatechin-3-gallate-induced stress signals in HT-29 human colon adenocarcinoma cells. Carcinogenesis 2003; 24: 1369–1378.

    Article  PubMed  Google Scholar 

  228. Kazi A, Smith DM, Zhong Q, Dou QP. Inhibition of bcl-x(l) phosphorylation by tea polyphenols or epigallocatechin-3-gallate is associated with prostate cancer cell apoptosis. Mol Pharmacol 2002; 62: 765–771.

    Article  PubMed  Google Scholar 

  229. Zhao Y, Yang LF, Ye M, Gu HH, Cao Y. Induction of apoptosis by epigallocatechin-3-gallate via mitochondrial signal transduction pathway. Prev Med 2004; 39: 1172–1179.

    Article  PubMed  Google Scholar 

  230. Hsu S, Lewis J, Singh B, et al. Green tea polyphenol targets the mitochondria in tumor cells inducing caspase 3-dependent apoptosis. Anticancer Res 2003; 23: 1533–1539.

    PubMed  Google Scholar 

  231. Dörrie J, Gerauer H, Wachter Y, Zunino SJ. Resveratrol induces extensive apoptosis by depolarizing mitochondrial membranes and activating caspase-9 in acute lymphoblastic leukemia cells. Cancer Res 2001; 61: 4731–4739.

    PubMed  Google Scholar 

  232. Mahyar-Roemer M, Katsen A, Mestres P, Roemer K. Resveratrol induces colon tumor cell apoptosis independently of p53 and precede by epithelial differentiation, mitochondrial proliferation and membrane potential collapse. Int J Cancer 2001; 94: 615–622.

    Article  PubMed  Google Scholar 

  233. Tinhofer I, Bernhard D, Senfter M, et al. Resveratrol, a tumor-suppressive compound from grapes, induces apoptosis via a novel mitochondrial pathway controlled by Bcl-2. FASEB J 2001; 15: 1613–1615.

    PubMed  Google Scholar 

  234. Mouria M, Gukovskaya AS, Jung Y, et al. Food-derived polyphenols inhibit pancreatic cancer growth through mitochondrial cytochrome c release and apoptosis. Int J Cancer 2002; 98: 761–769.

    Article  PubMed  Google Scholar 

  235. Luzi C, Brisdelli F, Cinque B, Cifone G, Bozzi A. Differential sensitivity to resveratrol-induced apoptosis of human chronic myeloid (K562) and acute lymphoblastic (HSB-2) leukemia cells. Biochem Pharmacol 2004; 68: 2019–2030.

    Article  PubMed  Google Scholar 

  236. Clement MV, Hirpara JL, Chawdhury SH, Pervaiz S. Chemopreventive agent resveratrol, a natural product derived from grapes, triggers CD95 signaling-dependent apoptosis in human tumor cells. Blood 1998; 92: 996–1002.

    PubMed  Google Scholar 

  237. Bernhard D, Tinhofer I, Tonko M, et al. Resveratrol causes arrest in the S-phase prior to Fas-independent apoptosis in CEM-C7H2 acute leukemia cells. Cell Death Differ 2000; 7: 834–842.

    Article  PubMed  Google Scholar 

  238. Notario B, Zamora M, Vinas O, Mampel T. All-trans-retinoic acid binds to and inhibits adenine nucleotide translocase and induces mitochondrial permeability transition. Mol Pharmacol 2003; 63: 224–231.

    Article  PubMed  Google Scholar 

  239. Rial E, Gonzáles-Barroso M, Fleury C, et al. Retinoids activate proton transport by the uncoupling proteins UPC1 and UPC2. EMBO J 1999; 18: 5827–5833.

    Article  PubMed  Google Scholar 

  240. Chomiki N, Voss JC, Warden CH. Structure-function relationships in UCP1, UCP2 and chimeras: EPR analysis and retinoic acid activation of UCP2. Eur J Biochem 2001; 268: 903–913.

    Article  PubMed  Google Scholar 

  241. Gianni M, Ponzanelli I, Mologni L, et al. Retinoid-dependent growth inhibition, differentiation and apoptosis in acute promyelocytic leukemia cells. Expression and activation of caspases. Cell Death Differ 2000; 7: 447–460.

    Article  PubMed  Google Scholar 

  242. Poot M, Hosier S, Swisshelm K. Distinct patterns of mitochondrial changes precede induction of apoptosis by all-trans-retinoic acid and N-(4-hydroxyphenyl)retinamide in MCF7 breast cancer cells. Exp Cell Res 2002; 279: 128–140.

    Article  PubMed  Google Scholar 

  243. Holmes WF, Soprano DR, Soprano KJ. Comparison of the mechanism of induction of apoptosis in ovarian carcinoma cells by the conformationally restricted synthetic retinoids CD437 and 4-HPR. J Cell Biochem 2003; 89: 262–278.

    Article  PubMed  Google Scholar 

  244. Holmes WF, Soprano DR, Soprano KJ. Elucidation of molecular events mediating induction of apoptosis by synthetic retinoids using a CD437-resistant ovarian carcinoma cell line. J Biol Chem 2002; 277: 45408–45419.

    Article  PubMed  Google Scholar 

  245. Asumendi A, Morales MC, Alvarez A, Arechaga J, Perez-Yarza G. Implication of mitochondria-derived ROS and cardiolipin peroxidation in N-(4-hydroxyphenyl)retinamide-induced apoptosis. Br J Cancer 2002; 86: 1951–1956.

    Article  PubMed  Google Scholar 

  246. Nicolaou KC, Pfefferkorn JA, Schuler F, Roecker AJ, Cao G-Q, Casida JE. Combinational synthesis of novel and potent inhibitors of NADH:ubiquinone oxidoreductase. Chem Biol 2000; 7: 979–992.

    Article  PubMed  Google Scholar 

  247. Wolvetang EJ, Johnson KL, Krauer K, Ralph SJ, Linnane AW. Mitochondrial respiratory chain inhibitors induce apoptosis. FEBS Lett 1994; 339: 40–44.

    Article  PubMed  Google Scholar 

  248. Yuki K, Miyauchi T, Kakinuma Y, et al. Mitochondrial dysfunction increases expression of endothelin-1 and induces apoptosis through caspase-3 activation in rat cardiomyocytes in vitro. J Cardiovasc Pharmacol 2000; 36: S205–208.

    PubMed  Google Scholar 

  249. Raghuvar Gopal DV, Narkar AA, Badrinath Y, Mishra KP, Joshi DS. Protection of Ewing’s sarcoma family tumor (ESFT) cell line SK-N-MC from betulinic acid induced apoptosis by α-DL-tocopherol. Toxicol Lett 2004; 153: 201–212.

    Article  PubMed  Google Scholar 

  250. Ehrhardt H, Fulda S, Fuhrer M, Debatin KM, Jeremias I. Betulinic acid-induced apoptosis in leukemia cells. Leukemia 2004; 18: 1406–1412.

    Article  PubMed  Google Scholar 

  251. Fulda S, Debatin KM. Betulinic acid induces apoptosis through a direct effect on mitochondria in neuroectodermal tumors. Med Pediatr Oncol 2000; 35: 616–618.

    Article  PubMed  Google Scholar 

  252. Fulda S, Scaffidi C, Susin SA, et al. Activation of mitochondria and release of mitochondrial apoptogenic factors by betulinic acid. J Biol Chem 1998; 273: 33942–33948.

    Article  PubMed  Google Scholar 

  253. Lapillonne H, Konopleva M, Tsao T, et al. Activation of peroxisome proliferator-activated receptor g by a novel synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid induces growth arrest and apoptosis in breast cancer cells. Cancer Res 2003; 63: 5926–5939.

    PubMed  Google Scholar 

  254. Dedov VN, Mandadi S, Armati PJ, Verkhratski A. Capsaicin-induced depolarization of mitochondria in dorsal root ganglion neurons is enhanced by vanilloid receptors. Neuroscience 2001; 103: 219–226.

    Article  PubMed  Google Scholar 

  255. Pal S, Choudhuri T, Chattopadhyay S, et al. Mechanisms of curcumin-induced apoptosis of Ehrlich’s ascites carcinoma cells. Biochem Biophys Res 2001; 288: 658–665.

    Article  Google Scholar 

  256. Pan MH, Chang WL, Lin-Shiau SY, Ho CT, Lin JK. Induction of apoptosis by garcinol and curcumin through cytochrome c release and activation of caspases in human leukemia HL-60 cells. J Agric Food Chem 2001; 49: 1464–1474.

    Article  PubMed  Google Scholar 

  257. Sundaram N, Pahwa AK, Ard MD, Lin N, Perkins E, Bowles AP, Jr. Selenium causes growth inhibition and apoptosis in human brain tumor cell lines. J Neurooncol 2000; 46: 125–133.

    Article  PubMed  Google Scholar 

  258. Zhong W, Oberley TD. Redox-mediated effects of selenium on apoptosis and cell cycle in the LNCaP human prostate cancer cell line. Cancer Res 2001; 61: 7071–7078.

    PubMed  Google Scholar 

  259. Jung U, Zheng X, Yoon SO, Chung AS. Se-methylselenocysteine induces apoptosis mediated by reactive oxygen species in HL-60 cells. Free Radic Biol Med 2001; 31: 479–489.

    Article  PubMed  Google Scholar 

  260. Kim TS, Jeong DW, Yun BY, Kim IY. Dysfunction of rat liver mitochondria by selenite: Induction of mitochondrial permeability transition through thiol-oxidation. Biochem Biophys Res 2002; 294: 1130–1137.

    Article  Google Scholar 

  261. Kim TS, Yun BY, Kim IY. Induction of the mitochondrial permeability transition by selenium compounds mediated by oxidation of the protein thiol groups and generation of the superoxide. Biochem Pharmacol 2003; 15: 2301–2311.

    Article  Google Scholar 

  262. Shilo S, Tirosh O. Selenite activates caspase-independent necrotic cell death in Jurkat T cells and J774.2 macrophages by affecting mitochondrial oxidant generation. Antioxid Redox Signal 2003; 5: 273–279.

    Article  PubMed  Google Scholar 

  263. Jiang C, Wang Z, Ganther H, Lu J. Caspases as key executors of methyl selenium-induced apoptosis (anoikis) of DU-145 prostate cancer cells. Cancer Res 2001; 61: 3062–3070.

    PubMed  Google Scholar 

  264. Neuzil J, Svensson I, Weber T, Weber C, Brunk UT. α-Tocopheryl succinate-induced apoptosis in Jurkat T cells involves caspase-3 activation, and both lysosomal and mitochondrial destabilization. FEBS Lett 1999; 445: 295-300.

    Article  PubMed  Google Scholar 

  265. Takahashi K, Loo G. Disruption of mitochondria during tocotrienol-induced apoptosis in MDA-MB-231 human breast cancer cells. Biochem Pharmacol 2004; 67: 315–324.

    Article  PubMed  Google Scholar 

  266. Yamamoto S, Tamai H, Ishisaka R, et al. Mechanism of alpha-tocopheryl succinate-induced apoptosis of promyelocytic leukemia cells. Free Radic Res 2000; 33: 407–418.

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to N. Hail Jr..

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hail, N. Mitochondria: A novel target for the chemoprevention of cancer. Apoptosis 10, 687–705 (2005). https://doi.org/10.1007/s10495-005-0792-8

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10495-005-0792-8

Keywords

Navigation