Skip to main content
Log in

Mechanism of fenretinide (4-HPR)-induced cell death

  • Published:
Apoptosis Aims and scope Submit manuscript

Abstract

4-HPR (fenretinide) is a synthetic analog of retinoic acid (RA) whose potential as a chemopreventative agent has gained support from in vitro and animal experiments and in limited clinical trials. Comparative analyses of cellular, biochemical, and molecular properties of fenretinide with RA using various tissue culture cells reveal that a key distinction between these two retinoids lies in the ability of fenretinide to induce programmed cell death, also known as apoptosis. Here we review the composite evidence for induction of apoptosis in fenretinide-treated cells. Assays used to validate apoptosis in various cell types are also summarized. Apoptosis in response to fenretinide primarily occurs by a receptor-independent mechanism, which is accompanied by increases in signaling molecules, e.g., ceramide, and cysteine-dependent aspartate-directed proteases, termed caspases, including execution caspase-3. Both caspase-3 inhibitor DEVD-CHO and ceramide synthase inhibitor fumonisin B1 (FB1) block fenretinide-induced apoptosis. Increase in caspase-3 appears to result from fenretinide-elicited stabilization of procaspase-3 zymogen. We also review apoptotic regulatory proteins such as inhibitor of apoptosis (IAPs) and second mitochondria-derived activator of caspase (SMACs) that participate in the coordinate control of caspase activities. The existence of a large number of proteins capable of modulating apoptosis via activation or inhibition of caspases, coupled with the fact that both the initiation and execution phases of apoptosis utilize pre-existing zymogens, which, once set in motion, culminates in an irreversible apoptotic cascade, raise the possibility that the on/off switch of apoptosis is linked to an intricate intracellular regulatory network, capable of responding to external stimuli such as fenretinide. This network functions to provide checks/balances of the need for apoptosis as well as to minimize and prevent untimely errors in apoptosis. We suggest that dynamic and coordinated regulation of apoptosis by such a hypothetical network in vivo may involve co-localization of pro- and anti-apoptotic proteins and their respective activators/inhibitors in a macromolecular modular unit which we propose to be named caspasomes. Fenretinide also induces apoptosis by elevating reactive oxygen species (ROS), unrelated to changes in ceramide-caspases. Thus multiple, distinct pathways contribute to the induction of apoptosis by fenretinide.

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. Ohshima M, Ward JM, Wenk ML. Preventive and enhancing effects of retinoids on the development of naturally occurring tumors of skin, prostate gland, and endocrine pancreas in aged male ACI/segHapBR rats. J Natl Cancer Inst 1985; 74: 517-524.

    Google Scholar 

  2. Pollard M, Luckert PH, Sporn MB. Prevention of primary prostate cancer in Lobund-Wistar rats by N-(4-hydroxyphenyl)retinamide. Cancer Res 1991; 51: 3610-3611.

    Google Scholar 

  3. Slawin K, Kadmon D, Park SH, et al. Dietary fenretinide, a synthetic retinoid, decreases the tumor incidence and the tumor mass of ras+myc-induced carcinomas in the mouse prostate reconstitution model system. Cancer Res 1993; 53: 4461-4465.

    Google Scholar 

  4. Ulukaya E, Wood EJ. Fenretinide and its relation to cancer. Cancer Treatment News 1999; 25: 229-235.

    Google Scholar 

  5. Modiano MR, Dalton WS, Lippman SM, Joffe L, Booth AR, Meyskens FL Jr. Phase II study of fenretinide (N-[4-hydroxyphenyl]retinamide) in advanced breast cancer and melanoma. Invest New Drugs 1990; 8: 317-319.

    Google Scholar 

  6. Evans TRJ, Kaye SB. Retinoids: Present role and future potential. Br J Cancer 1999; 80: 1-8.

    Google Scholar 

  7. Lippman SM, Lotan R. Advances in the development of retinoids as chemopreventive agents. J Nutr 2000; 130: 479S-482S.

    Google Scholar 

  8. Zhang D, Holmes WF, Wu S, Soprano DR, Soprano KJ. Retinoids and ovarian cancer. J Cell Physiol 2000; 185: 1-20.

    Google Scholar 

  9. Torrisi R, Mezzeth M, Johansson H, et al. Time course of fenretinide-induced modulation of circulating insulin-like growth factor (IGF)-1, IGF-II and IGFBP-3 in a bladder cancer chemoprevention trial. Int J Cancer 2000; 87: 601-605.

    Google Scholar 

  10. Kurie J, Lee JS, Khuri FR, et al. N-(4-hydroxyphenyl)retinimide in the chemoprevention of squamous metaplasia and dysplasia of the bronchial epithelium. Clin Cancer Res 2000; 6: 2973-2979.

    Google Scholar 

  11. Cobleigh MA, Dowlatshahi K, Deutsch TA, et al. Phase I/II trial of tamoxifen with or without fenretinide, an analog of vitamin A, in women with metastatic breast cancer. J Clin Oncol 1993; 11: 474-477.

    Google Scholar 

  12. Chodak GW, Rukstalis D, Kellman HM, Williams M. Phase II study of the retinoid analogue 4-HPR in men with carcinoma of the prostate. J Urol (Suppl) 1993; 149: 257A.

    Google Scholar 

  13. Cassano E, Coopmans de Yoldi G, Ferranti C, et al. Mammographic patterns in breast cancer chemoprevention with fenretinide (4-HPR). Eur J Cancer 1993; 29A: 2161-2163.

    Google Scholar 

  14. Hong WK, Lippman SM, Itri LM, et al. Prevention of second primary tumors with isotretinoin in squamous-cell carcinoma of the head and neck. N Engl J Med 1990; 323: 795-801.

    Google Scholar 

  15. Pienta KJ, Esper PS, Zwas F, Krzeminski R, Flaherty LE. Phase II chemoprevention trial of oral fenretinide in patients at risk for adenocarcinoma of the prostate. Am J Clin Oncol 1997; 20: 36-39.

    Google Scholar 

  16. Mehta RC, Moon RC, Hawthorne M, Formelli F, Costa A. Distribution of fenretinide in the mammary gland of breast cancer patient. Eur J Cancer 1991; 27: 138-141.

    Google Scholar 

  17. Modiano MR, Peng YM, Xu MJ, et al. Chemoprevention of breast cancer with fenretinide (4-HPR): Data from benign and malignant breast tissue and plasma 4-HPR kinetics. Proc Am Assoc Cancer Res 1993; 34: A3290.

    Google Scholar 

  18. Swanson BN, Zaharevitz DW, Sporn MB. Pharmacokinetics of N-(4-hydroxyphenyl)-all-trans-retinamide in rats. Drug Metab Dispos 1980; 8: 168-172.

    Google Scholar 

  19. Rotmensz N, De Palo G, Formelli F, et al. Long-term tolerability of fenretinide (4-HPR) in breast cancer patients. Eur J Cancer 1991; 27: 1127-1131.

    Google Scholar 

  20. Gross EG, Peck GL, DiGiovanna JJ. Adverse reaction to fenretinide, a synthetic retinoid [letter]. Arch Dermatol 1991; 127: 1849-1850.

    Google Scholar 

  21. De Urquiza AM, Liu S, Sjoberg M, et al. Docosahexaenoic acid, a ligand for the retinoid X receptor in mouse brain. Science 2000; 290: 2140-2144.

    Google Scholar 

  22. Sabichi AL, Hendricks DT, Bober MA, Birrer MJ. Retinoic acid receptor beta expression and growth inhibition of gynecologic cancer cells by the synthetic retinoid N-(4-hydroxyphenyl) retinamide. J Natl Cancer Inst 1998; 90: 597-605.

    Google Scholar 

  23. Liu G, Wu M, Levi G, Ferrari N. Inhibition of cancer cell growth by all-trans retinoic acid and its analog N-(4-hydroxyphenyl) retinamide: A possible mechanism of action via regulation of retinoid receptors expression. Int J Cancer 1998; 78: 248-254.

    Google Scholar 

  24. Fanjul AN, Delia D, Pierotti MA, et al. 4-Hydroxyphenyl retinamide is a highly selective activator of retinoid receptors. J Biol Chem 1996; 271: 22441-22446.

    Google Scholar 

  25. Sun SY, 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.

    Google Scholar 

  26. Lovat PE, Ranalli M, Annichiarrico-Petruzelli M, et al. Effector mechanisms of fenretinide-induced apoptosis in neuroblastoma. Exp Cell Res 2000; 260: 50-60.

    Google Scholar 

  27. Sheikh MS, Shao ZM, Li XS, et al. N-(4-hydroxyphenyl)retinamide (4-HPR)-mediated biological actions involve retinoid receptor-independent pathways in human breast carcinoma. Carcinogenesis 1995; 16: 2477-2486.

    Google Scholar 

  28. Sani BP, Shealy YF, Hill DL. N-(4-hydroxyphenyl)retinamide: Interactions with retinoid-binding proteins/receptors. Carcinogenesis 1995; 16: 2531-2534.

    Google Scholar 

  29. 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.

    Google Scholar 

  30. Torrisi R, Parodi S, Fontana V, et al. Effect of fenretinide on plasma IGF-I and IGFBP-3 in early breast cancer patients. Int J Cancer 1998; 76: 787-790.

    Google Scholar 

  31. Clifford JL, Menter DG, Wang M, Lotan R, Lippman SM. Retinoid receptor-dependent and-independent effects of N-(4-hydroxyphenyl)retinamide in F9 embryonal carcinoma cells. Cancer Res 1999; 59: 14-18.

    Google Scholar 

  32. Di Vinci A, Geido E, Infusini E, Giaretti W. Neuroblastoma cell apoptosis induced by the synthetic retinoid N-(4-hydroxyphenyl)retinamide. Int J Cancer 1994; 59: 422-426.

    Google Scholar 

  33. Springer LN, Stewart BW. N-(4-hydroxyphenyl)retinamide-induced death in human lymphoblastoid cells: 50 kb DNA breakage as a means of distinguishing apoptosis from necrosis. Cancer Lett 1998; 128: 189-196.

    Google Scholar 

  34. Igawa M, Tanabe T, Chodak GW, Rukstalis DB. N-(4-hydroxyphenyl) retinamide induces cell cycle specific growth inhibition in PC-3 cells. Prostate 1994; 24: 299-305.

    Google Scholar 

  35. Hsieh TC, Wu JM. Effects of fenretinide (4-HPR) on prostate LNCaP cell growth, apoptosis, and prostate-specific gene expression. Prostate 1997; 33: 97-104.

    Google Scholar 

  36. Jinno H, Steiner MG, Mehta RG, Osborne MP, Telang NT. Inhibition of aberrant proliferation and induction of apoptosis in HER-2/neu oncogene transformed human mammary epithelial cells by N-(4-hydroxyphenyl)retinamide. Carcinogenesis 1999; 20: 229-236.

    Google Scholar 

  37. DiPietrantonio AM, Hsieh TC, Olson SC, Wu JM. Regulation of G1/S transition and induction of apoptosis in HL-60 leukemia cells by fenretinide (4HPR). Int J Cancer 1998; 78: 53-61.

    Google Scholar 

  38. Chellappan SP, Hiebert S, Mudryj M, Horowitz JM, Nevins JR. The E2F transcription factor is a cellular target for the RB protein. Cell 1991; 65: 1053-1061.

    Google Scholar 

  39. Hamel PA, Gill RM, Phillips RA, Gillie BL. Transcriptional repression of the E2-containing promoters EIIaE, c-myc, and RB1 by the product of the RB1 gene. Mol Cell Biol 1992; 12: 3431-3438.

    Google Scholar 

  40. Clarke PG, Clarke S. Nineteenth century research on naturally occurring cell death and related phenomena. Anat Embryol (Berl) 1996; 193: 81-99.

    Google Scholar 

  41. Jacobson MD, Weil M, Raff MC. Programmed cell detah in animal development. Cell 1997, 88: 347-354.

    Google Scholar 

  42. Lockshin RA, Williams CM. Programmed cell death. II. Endocrine potentiation of the breakdown of the intersegmental muscle of silkmoths. J Insect Physiol 1964; 10: 643.

    Google Scholar 

  43. Kerr JF, Wyllie AH, Currie AR. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 1972; 26: 239-257.

    Google Scholar 

  44. Kerr JF, Searle J. A mode of cell loss in malignant neoplasms. J Pathol 1972; 106: Pxi

    Google Scholar 

  45. Thompson CB. Apoptosis in the pathogenesis and treatment of disease. Science 1995; 267: 1456-1462.

    Google Scholar 

  46. Cotman CW, Anderson AJ. A potential role for apoptosis in neurodegeneration and Alzheimer's disease. Mol Neurobiol 1995; 10: 19-45.

    Google Scholar 

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

    Google Scholar 

  48. Famularo G, De Simone C, Marcellini S. Apoptosis: Mechanisms and relation to AIDS. Med Hypotheses 1997; 48: 423-429.

    Google Scholar 

  49. Wyllie AH, Kerr JF, Currie AR Cell death: The significance of apoptosis. Int Rev Cytol 1980; 68: 251-306.

    Google Scholar 

  50. Arends MJ, Wyllie AH. Apoptosis: Mechanisms and roles in pathology. Int Rev Exp Pathol 1991; 32: 223-254.

    Google Scholar 

  51. McConkey DJ. Cellular signaling in cell death. New Horiz 1993; 1: 52-59.

    Google Scholar 

  52. Martin SJ, Reutelingsperger CP, McGahon AJ, van Schie RC, LaFace DM, Green DR. Early redistribution of plasma membrane phosphatidyllserie is a general feature of apoptosis regardless of the initiating stimulus: Inhibition by overexpression of bcl-2 and Abl. J Exp Med 1995; 182: 1545-1556.

    Google Scholar 

  53. Wyllie AH. Glucocorticoid-induced thymocyte apoptosis is associated with endogenous endonuclease activation. Nature 1980; 284: 555-556.

    Google Scholar 

  54. Wyllie AH, Morris RG. Hormone-induced cell death. Purification and properties of thymocytes undergoing apoptosis after glucocorticoid treatment. Am J Pathol 1982; 109: 78-87.

    Google Scholar 

  55. Kaufmann SH. Induction of endonucleolytic DNA cleavage in human acute myelogenous leukemia cells by etoposide, camptothecin, and other cytotoxic anticancer drugs: A cautionary note. Cancer Res 1989; 49: 5870-5878.

    Google Scholar 

  56. Lazebnik YA, Kaufmann SH, Desnoyers S, Poirier GG, Earnshaw WC. Cleavage of poly(ADP-ribose) polymerase by a proteinase with properties like ICE. Nature 1994; 371: 346-347.

    Google Scholar 

  57. Lazebnik YA, Cole S, Cooke CA, Nelson WG, Earnshaw WC. Nuclear events of apoptosis in vitro in cell-free mitotic extracts: A model system for analysis of the active phase of apoptosis. J Cell Biol 1993; 123: 7-22.

    Google Scholar 

  58. Vaux DL. Caspases and apoptosis-biology and terminology. Cell Death & Differen 1999; 6: 493-494.

    Google Scholar 

  59. McDonnell TJ, Deane N, Platt FM, et al. Bcl-2-immunoglobulin transgenic mice demonstrate extended B cell survival and follicular lymphoproliferation. Cell 1989; 57: 79-88.

    Google Scholar 

  60. Tsujimoto Y. Overexpression of the human BCL-2 gene product results in growth enhancemen of Epstein Barr virus-immortalized B cells. Proc Natl Acad Sci USA 1989; 86: 1958-1962.

    Google Scholar 

  61. Hockenbery D, Nunez G, Milliman C, Schreiber RD, Korsmeyer SJ. Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature 1990; 348: 334-336.

    Google Scholar 

  62. Boise LH, Gonzalez Garcia M, Postema CE, et al. Bcl-x, a bcl-2-related gene that functions as a dominant regulator of apoptotic cell death. Cell 1993; 74: 597-608.

    Google Scholar 

  63. Reynolds JE, Yang T, Qian L, et al. Mcl-1, a member of the Bcl-2 family, delays apoptosis induced by c-Myc overexpression in Chinese hamster ovary cells. Cancer Res 1994; 54: 6348-6352.

    Google Scholar 

  64. Oltvai ZN, Milliman CL, Korsmeyer SJ. Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell 1993; 74: 609-619.

    Google Scholar 

  65. McDonnell TJ, Beham A, Sarkiss M, Andersen MM, Lo P. Importance of the Bcl-2 family in cell death regulation. Experientia 1996; 52: 1008-1017.

    Google Scholar 

  66. Chittenden T, Harrington EA, O'Connor R, et al. Induction of apoptosis by the Bcl-2 homologue Bak. Nature 1995; 374: 733-736.

    Google Scholar 

  67. Farrow SN, White JH, Martinou I, et al. Cloning of a bcl-2 homologue by interaction with adenovirus E1B 19K. Nature 1995; 374: 731-733.

    Google Scholar 

  68. Kiefer MC, Brauer MJ, Powers VC, et al. Modulation of apoptosis by the widely distributed Bcl-2 homologue Bak. Nature 1995; 374: 736-739.

    Google Scholar 

  69. Yuan J, Shaham S, Ledoux S, Ellis HM, Horvitz HR. The C. elegans cell death gene ced-3 encodes a protein similar to mammalian interleukin-1 beta-converting enzyme. Cell 1993; 75: 641-652.

    Google Scholar 

  70. Alnemri ES, Livingston DJ, Nicholson DW, et al. Human ICE/CED-3 protease nomenclature [letter]. Cell 1996; 87: 171.

    Google Scholar 

  71. Di Vinci A, Geido E, Infusani E, Giaretti W. Neuroblastoma cell apoptosis induced by the synthetic retinoid N-(4-hydroxyphenyl)retinamide. Int J Cancer 1994; 59: 422-426.

    Google Scholar 

  72. Mariorri A, Marcora E, Bunone G, et al. N-(4-hydroxyphenyl) retinamide: A potent inducer of apoptosis in human neuroblastoma cells. J Natl Cancer Inst 1994; 86: 1245-1247.

    Google Scholar 

  73. Hsieh TC, Ng C, Wu JM. The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) exerts antiproliferative and apoptosis-inducing effects in the androgen-independent human prostatic JCA-1 cells. Biochem Mol Biol Intl 1995; 37: 499-506.

    Google Scholar 

  74. Delia D, Aiello A, Formelli F, et al. Regulation of apoptosis induced by the retinoid N-(4-hydroxyphenyl)retinamide and effect of deregulated bcl-2. Blood 1995; 85: 359-367.

    Google Scholar 

  75. Ponzoni M, Bocca P, Chiesa V, et al. Differential effects of N-(4-hydroxyphenyl)retinamide and retinoic acid on neuroblastoma cells: Apoptosis versus differentiation. Cancer Res 1995; 55: 853-861.

    Google Scholar 

  76. Sheikh MS, Shao ZM, Li XS, et al. N-(4-hydroxyphenyl) retinamide (4-HPR)-mediated biological actions involve retinoid receptor-independent pathways in human breast carcinoma. Carcinogenesis 1995; 16: 2477-2486.

    Google Scholar 

  77. Pellegrini R, Mariotti A, Tagliabue E, et al. Modulation of markers associated with aggressiveness in human breast cancer cell lines by N-(4-hydroxyphenyl)retinamide. Cell Growth & Differentiation 1995; 6: 863-869.

    Google Scholar 

  78. Supino A, Crosti M, Clerici M, et al. Induction of apoptosis by fenretinide (4HPR) in human ovarian carcinoma cells and its association with retinoic acid receptor expression. Int J Cancer 1996; 65: 491-497.

    Google Scholar 

  79. DiPietrantonio AM, Hsieh TC, Wu JM. Differential effects of retinoic acid (RA) and N-(4-hydroxyphenyl)retinamide (4-HPR) on cell growth, induction of differentiation, and changes in p34cdc2, bcl-2, and actin expression in the human promyelocytic HL-60 leukemic cells. Biochem Biophys Res Commun 1996; 224: 837-842.

    Google Scholar 

  80. Fanjul AN, Delia D, Pierotti MA, Rideout D, Qiu J, Pfahl M. N-(4-hydroxy-phenyl)retinamide is a highly selective activator of retinoid receptors. J Biol Chem 1996; 271: 22441-22446.

    Google Scholar 

  81. Wang TTY, Phang JM. Effect of N-(4-hydroxyphenyl) retinamide on apoptosis in human breast cancer cells. Cancer Lett 1996; 107: 65-71.

    Google Scholar 

  82. 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.

    Google Scholar 

  83. 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.

    Google Scholar 

  84. Hsieh TC, Wu JM. Effects of fenretinide (4-HPR) on prostate LNCaP cell growth, apoptosis, and prostate-specific gene expression. The Prostate 1997; 33: 97-104.

    Google Scholar 

  85. 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.

    Google Scholar 

  86. Robertson KM, Penland SN, Padilla GM, et al. Fenretinide: Induction of apoptosis and endogenous transforming growth factor ? in PC-3 prostate cancer cells. Cell Growth & Differentiation 1997; 8: 101-111.

    Google Scholar 

  87. Chan LL, Zhang S, Shao J, Waikel R, Thompson EA, Chan TS. N-(4-hydroxyphenyl)retinamide induces apoptosis in T lymphoma and T lymphoblastoid leukemic cells. Leukemia & Lymphoma 1997; 25: 271-280.

    Google Scholar 

  88. Springer LN, Stewart BW. N-(4-hydroxyphenyl)retinamide-induced death in human lymphoblastoid cells: 50 kb DNA breakage as a means of distinguishing apoptosis from necrosis. Cancer Lett 1998; 128: 189-196.

    Google Scholar 

  89. Sabichi AL, Hendricks DT, Bober MA, Birrer MJ. Retinoic acid receptor ? expression and growth inhibition of gynecologic cancer cells by the synthetic retinoid N-(4-hydroxyphenyl)retinamide. J Natl Cancer Inst 1998; 90: 597-605.

    Google Scholar 

  90. Liu G, Wu M, Levi G, Ferrari N. Inhibition of cancer cell growth by all-trans-retinoic acid and its analog N-(4-hydroxyphenyl)retinamide: A possible mechanism of action via regulation of retinoid receptors expression. Int J Cancer 1998; 78: 248-254.

    Google Scholar 

  91. Saitoh Y, Goto T, Puduvalli V, et al. Induction of apoptosis by N-(4-hydroxyphenyl)retinamide. Int J Oncol 1999; 15: 499-504.

    Google Scholar 

  92. Clifford JL, Menter DG, Wang M, Lotan R, Lippman SM. Retinoid receptor-dependent and-independent effects of N-(4-hydroxyphenyl)retinamide in F9 embryonal carcinoma cells. Cancer Res 1998; 59: 14-18.

    Google Scholar 

  93. DiPietrantonio AM, Hsieh TC, Olson SC, Wu JM. Altered expression of the G1/S checkpoint regulators (cyclin D1, cdk4, pRB) and transient modulation of poly(ADP-ribose) polymerase and ceramide level changes by fenretinide N-(4-hydroxyphenyl)retinamide, 4-HPR) in human promyelocytic leukemic cells. Int J Cancer 1998; 78: 53-61.

    Google Scholar 

  94. Maurer BJ, Metelitsa LS, Seeger RC, Cabot MC, Reynolds CP. Increase 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.

    Google Scholar 

  95. Jinno H, Steiner MG, Mehta RG, Osborne MP, Telang NT. Inhibition of aberrant proliferation and induction of apoptosis in HER-2/neu oncogene transformed human mammary epithelial cells by N-(4-hydroxyphenyl)retinamide. Carcinogenesis 1999; 20: 229-236.

    Google Scholar 

  96. Puduvalli VK, Saito Y, Xu R, Kouraklis GP, Levin VA, Kyritsis AP. Fenretinide activates caspases and induces apoptosis in gliomas. Clin Cancer Res 1999; 5: 2230-2235.

    Google Scholar 

  97. Kitareewan S, Spinella MJ, Allopenna J, Reczek PR, Dmitrovsky E. 4HPR triggers apoptosis but not differentiation in retinoid sensitive and resistant human embryonal carcinoma cells through an RAR? independent pathway. Oncogene 1999; 18: 5747-5755.

    Google Scholar 

  98. Sun SY, 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.

    Google Scholar 

  99. Sun SY, Li W, Yue P, Lippman SM, Hong WK, Lotan R. Mechanism of N-(4-hydroxyphenyl)retinamide-induced apoptosis in human cancer cells by different mechanisms. Cancer Res 1999; 59: 2493-2498.

    Google Scholar 

  100. Montaldo PG, Pagnan G, Pastorino F, et al. N-(4-hydroxyphenyl)retinamide is cytotoxic to melanoma cells in vitro through induction of programmed cell death. Int J Cancer 1999; 81: 262-267.

    Google Scholar 

  101. Webber MM, Bello-DeOcampo D, Quader S, Deocampo ND, Metcalfe WS, Sharp RM. Modulation of the malignant phenotype of human prostate cancer cells by N-(4-hydroxyphenyl)retinamide (4-HPR). Clin & Exp Metastasis 1999; 17: 255-263.

    Google Scholar 

  102. Yang X, Hao Y, Ding Z, Pater A. BAG-1 promotes apoptosis induced by N-(4-hydroxyphenyl)retinamide in human cervical carcinoma cells. Exp Cell Res 2000; 256: 491-499.

    Google Scholar 

  103. DiPietrantoniuo AM, Hsieh TC, Juan G, Traganos F, Darzynkiewicz Z, Wu JM. Fenretinide-induced caspase 3 activity involves increased protein stability in a mechanism distinct from reactive oxygen species elevation. Cancer Res 2000; 60: 4331-4335.

    Google Scholar 

  104. Lovat PE, Ranalli M, Annichiarrico-Petruzzelli M, et al. Effector mechanisms of fenretinide-induced apoptosis in neuroblastoma. Exp Cell Res 2000; 260: 50-60.

    Google Scholar 

  105. 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.

    Google Scholar 

  106. Kalemkerian GP, Slusher R, Ramalingam S, Gadgeel S, Mabry M. Growth inhibition and induction of apoptosis by fenretinide in small-cell lung cancer cell lines. J Natl Cancer Inst 1995; 87: 1674-1680.

    Google Scholar 

  107. Zou C, Wang L, Liebert M, Grossman HB, Lotan R, Wei Q. Combined effect of chemopreventative agent N-(4-hydroxyphenyl) retinamide (4-HPR) and gamma-radiation on bladder cancer cell lines. Int J Oncol 1998; 13: 1037-1041.

    Google Scholar 

  108. Bedner A, Li X, Gorczyca W, Melamed MR, Darzynkewicz Z. Analysis of apoptosis by laser scanning cytometry. Cytometry 1999; 35: 181-195.

    Google Scholar 

  109. Obeid LM, Linardic CM, Karolak LA, Hannun YA. Programmed cell death induced by ceramide. Science 1993; 259: 1769-1771.

    Google Scholar 

  110. Cifone MG, De Maria R, Roncaioli P, et al. Apoptotic signaling through CD95 (Fas/Apo-1) activates an acidic sphingomyelinase. J Exp Med 1994; 180: 1547-1552.

    Google Scholar 

  111. Kuroki J, Hirokawa M, Kitabayashi A, et al. Cell-permeable ceramide inhibits the growth of B lymphoma Raji cells lacking TNF-alpha-receptors by inducing G0/G1 arrest but not apoptosis: A new model for dissecting cell-cycle arrest and apoptosis. Leukemia 1996; 10: 1950-1958.

    Google Scholar 

  112. Mizushima N, Koike R, Kohsaka H, et al. Ceramide induces apoptosis via CPP32 activation. FEBS Lett 1996; 395: 267-271.

    Google Scholar 

  113. Dobrowsky RT, Kamibayashi C, Mumby MC, Hannun YA. Ceramide activates heterotrimeric protein phosphatase 2A. J Biol Chem 1993; 268: 15523-15530.

    Google Scholar 

  114. Joseph CK, Byun HS, Bittman R, Kolesnick RN. Substrate recognition by ceramide-activated protein kinase. Evidence that kinase activity is proline-directed. J Biol Chem 1993; 268: 20002-20006.

    Google Scholar 

  115. Yang Z, Costanzo M, Golde DW, Kolesnick RN. Tumor necrosis factor activation of the sphingomyelin pathway signals nuclear factor kappa B translocation in intact HL-60 cells. J Biol Chem 1993; 268: 20520-20523.

    Google Scholar 

  116. Sawai H, Okazaki T, Takeda Y, et al. Ceramide-induced translocation of protein kinase C-delta and-epsilon to the cytosol. Implications in apoptosis. J Biol Chem 1997; 272: 2452-2458.

    Google Scholar 

  117. Larrick JW, Wright SC. Cytotoxic mechanism of tumor necrosis factor-alpha. FASEB J 1990; 4: 3215-3223.

    Google Scholar 

  118. Manome Y, Datta R, Taneja N, et al. Coinduction of c-jun gene expression and internucleosomal DNA fragmentation by ionizing radiation. Biochemistry 1993; 32: 10607-10613.

    Google Scholar 

  119. Mayer M, Noble M. N-acetyl-l-cysteine is a pluripotent protector against cell death and enhancer of trophic factor-mediated cell survival in vitro. Proc Natl Acad Sci USA 1994; 91: 7496-7500.

    Google Scholar 

  120. Cossarizza A, Franceschi C, Monti D, et al. Protective effect of N-acetylcysteine in tumor necrosis factor-alpha-induced apoptosis in U937 cells: The role of mitochondria. Exp Cell Res 1995; 220: 232-240.

    Google Scholar 

  121. Buttke TM, Sandstrom PA. Oxidative stress as a mediator of apoptosis. Immunol Today 1994; 15: 7-10.

    Google Scholar 

  122. Dumont A, Hehner SP, Hofmann TG, Ueffing M, Droge W, Schmitz ML. Hydrogen peroxide-induced apoptosis is CD95-independent, requires the release of mitochondria-derived reactive oxygen species and the activation of NF-kappaB. Oncogene 1999; 18: 747-757.

    Google Scholar 

  123. Kamata H, Hirata H. Redox regulation of cellular signalling. Cell Signal 1999; 11: 1-14.

    Google Scholar 

  124. Zheng M, Storz G. Activation of the OxyR transcription factor by reversible disulfide bond formation. Science 1998; 279: 1718-1721.

    Google Scholar 

  125. Reed JC. Bcl-2 and the regulation of programmed cell death. J Cell Biol 1994; 124: 1-6.

    Google Scholar 

  126. Cohen GM. Caspases: The execution of apoptosis. Biochem J 1997; 326: 1-16.

    Google Scholar 

  127. Shen JC, Wang TT, Chang S, Hursting SD. Mechanistic studies of the effects of the retinoid N-(4-hydroxyphenyl)retinamide on prostate cancer cell growth and apoptosis. Mol Carcinogenesis 1999; 24: 160-168.

    Google Scholar 

  128. Orth K, O'Rouke K, Salvesen GS, Dixit VM. Molecular ordering of apoptotic mammalian CED-3/ICE-like proteases. J Biol Chem 1996; 271: 20977-20980.

    Google Scholar 

  129. Chen YR, Zhou G, Tan TH. C-Jun N-terminal kinase modulates apoptotic signaling induced by N-(4-hydroxyphenyl) retinamide. Mol Pharmacol 1999; 56: 1271-1279.

    Google Scholar 

  130. Ellis HM, Horvitz HR. Genetic control of programmed cell death in the nematode C. elegans. Cell 1986; 44: 817-829.

    Google Scholar 

  131. Song Z, Ste1ler H. Death by design: Mechanism and control of apoptosis. Trends Cell Biol 1999; 9: M49-M52.

    Google Scholar 

  132. Yuan J, Shaham S, Ledoux S, Ellis HM, Horvitz HR. The C. elegans cell death gene ced-3 encodes a protein similar to mammalian interleukin-1 beta-converting enzyme. Cell 1993; 75: 641-652.

    Google Scholar 

  133. Earnshaw WC, Martinis LM, Kaufmann SH. Mammalian caspases: Structure, activation, substrates, and functions during apoptosis. Annu Rev Biochem 1999; 68: 383-424.

    Google Scholar 

  134. Thomberry NA, Lazebnik Y. Caspases: Enemies within. Science 1998; 281: 1312-1316.

    Google Scholar 

  135. Thornberry NA, Rano TA, Peterson EP, et al.A combinational approach defines specificies of members of the caspase family and granzyme B. Functional relationships established for key mediators of apoptosis. J Biol Chem 1997; 272: 17907-17911.

    Google Scholar 

  136. Sun XM, MacFarlane M, Zhuang J, Wolf BB, Green DR, Cohen GM. Distinct caspase cascades are initiated in receptor-mediated and chemica-induced apoptosis. J Biol Chem 1999; 274: 5053-5060.

    Google Scholar 

  137. Wilson MR. Apoptotic signal transduction: Emerging pathways. Biochem Cell Biol 1998; 76: 573-582.

    Google Scholar 

  138. Fesik SW. Insights into programmed cell death through structural biology. Cell 2000; 103: 273-282.

    Google Scholar 

  139. Coffey RNT, Watson RWG, Fitzpatrick JM. Signaling for the caspases: Their role in prostate cell apoptosis. J Urol 2001; 165: 5-14.

    Google Scholar 

  140. Bossy-Wetzel E, Green DR. Apoptosis: Checkpoint at the mitochondrial frontier. Mutat Res 1999; 434: 243-251.

    Google Scholar 

  141. Wolf BB, Schuler M, Echeverri F, Green DR. Caspase-3 is the primary activator of apoptotic DNA fragmentation via DNA fragmentation factor-45/inhibitor of caspase-activated DNase inactivation. J Biol Chem 1999; 274: 30651-30656.

    Google Scholar 

  142. Voe1ke1-Johnson C, Entingh AJ, Wold WS, Gooding LR, Laster SM. Activation of intracellular proteases is an early event in TNF-induced apoptosis. J Immunol 1995; 154: 1707-1716.

    Google Scholar 

  143. Widmann C, Gerwins P, Johnson NL, Jarpe MB, Johnson GL. MEK kinase 1, a substrate for DEVD-directed caspases, is involved in genotoxin-induced apoptosis. Mol Cell Biol 1998; 18: 2416-2429.

    Google Scholar 

  144. Widmann C, Gibson S, Johnson GL. Caspase-dependent cleavage of signaling proteins during apoptosis. J Biol Chem 1998; 273: 7141-7147.

    Google Scholar 

  145. Rao L, Perez D, White E. Lamin proteolysis facilitates nuclear events during apoptosis. J Cell Biol 1996; 135: 1441-1455.

    Google Scholar 

  146. Ku NO, Liao J, Omary MB. Apoptosis generates stable fragments of human type I keratins. J Biol Chem 1997; 272: 33197-33203.

    Google Scholar 

  147. Swanton E, Bishop N, Woodman P. Human rabaptin-5 is selectively cleaved by caspase-3 during apoptosis. J Biol Chem 1999; 274: 37583-37590.

    Google Scholar 

  148. Mashima T, Naito M, Fujita N, Noguchi K, Tsuruo T. Identification of actin as a substrate of ICE and an ICE-like protease and involvement of an ICE-like protease but not ICE in VP-16-induced U937 apoptosis. Biochem Biophys Res Commun 1995; 217: 1185-1192.

    Google Scholar 

  149. Ravi R, Bedi A, Fuchs EJ. CD95 (Fas)-induced caspasemediated proteolysis of NF-?B. Cancer Res 1998; 58: 882-886.

    Google Scholar 

  150. Barkett M, Xue D, Horvitz HR, Gilmore TD. Phosphorylation of I?B-? inhibits its cleavage by caspase CPP32 in vitro. J Biol Chem 1997; 272: 29419-29422.

    Google Scholar 

  151. Wang X, Zelenski NG, Yang J, Sakai J, Brown MS, Goldstein JL. Cleavage of sterol regulatory element binding proteins (SREBPs) by CPP32 during apoptosis. EMBO J 1996; 15: 1012-1020.

    Google Scholar 

  152. Jacobson MK, Jacobson EL. Discovering new ADP-ribose polymer cycles: Protecting the genome and more. Trends Biochem Sci 1999; 24: 415-417.

    Google Scholar 

  153. Simbulan-Rosenthal CM, Rosenthal DS, Iyer S, Boulares AH, Smulson ME. Transient poly(ADP-ribosyl)ation of nuclear proteins and role of poly(ADP-ribose)polymerase in the early stages of apoptosis. J Biol Chem 1998; 273: 13703-13712.

    Google Scholar 

  154. Deveraux QL, Reed JC. IAP family proteins-Suppressors of apoptosis. Genes & Development 1999; 13: 239-252.

    Google Scholar 

  155. Deveraux QL, Leo E, Stennicke HR, Welsh K, Salvesen GS, Reed JC. Cleavage of human inhibitor of apoptosis protein XIAP results in fragments with distinct specificities for caspases. EMBO J 1999; 18: 5242-5251.

    Google Scholar 

  156. Huang Q, Deveraux QL, Maeda S, et al. Evolutionary conservation of apoptosis mechanisms: Lepidoopteran and baculoviral inhibitor of apoptosis proteins are inhibitors of mammalian caspase-9. Proc Natl Acad Sci USA 2000; 97: 1427-1432.

    Google Scholar 

  157. Du C, Fang M, Li Y, Li L, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000; 102: 33-42.

    Google Scholar 

  158. Green DR. Apoptotic pathways: Paper wraps stone blunts scissors. Cell 2000; 102: 1-4.

    Google Scholar 

  159. Chau BN, Cheng EH, Kerr DA, Hardwick JM. Aven, a novel inhibitor of caspase activation, binds bcl-xL and Apaf-1. Mol Cell 2000; 6: 31-40.

    Google Scholar 

  160. Inohara N, Ding L, Chen S, Nunez G. Harakiri, a novel regulator of cell death, encodes a protein that activates apoptosis and interacts selectively with survival-promoting proteins Bcl-2 and Bcl-xL. EMBO J 1997; 16: 1686-1694.

    Google Scholar 

  161. Lee JH, Takahashi T, Yasuhara N, Inazawa J, Kamada S, Tsujimoto Y. Bis, a Bcl-2-binding protein that synergizes with Bcl-2 in preventing cell death. Oncogene 1999; 18 6183-6190.

    Google Scholar 

  162. Zhang H, Xu Q, Krajewski S, et al. BAR: An apoptosis regulator at the intersection of caspases and Bcl-2 family proteins. Proc Natl Acad Sci USA 2000; 97: 2597-2602.

    Google Scholar 

  163. Cardone MH, Roy N, Stennicke HR, et al. Regulation of cell death protease caspase-9 by phosphorylation. Science 1998; 282: 1318-1321.

    Google Scholar 

  164. Halder S, Jena N, Croce CM. Inactivation of Bcl-2 by phosphorylation. Proc Natl Acad Sci USA 1995; 92: 4507-4511.

    Google Scholar 

  165. Bedner E, Smolewski P, Amstad P, Darzynkiewicz Z. Activation of caspases measured in situ by binding of fluorochrome-labeled inhibitors of caspases (FLICA): Correlation with DNA fragmentation. Exp Cell Res 2000; 260: 308-313.

    Google Scholar 

  166. Smolewski P, Bedner E, Du L, et al. Detection of caspase activation by fluorochrome-labeled inhibitors of caspases (FLICA). Multiparameter analysis by laser scanning cytometry. Cytometry 2001; 44: 73-82.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wu, J.M., DiPietrantonio, A.M. & Hsieh, TC. Mechanism of fenretinide (4-HPR)-induced cell death. Apoptosis 6, 377–388 (2001). https://doi.org/10.1023/A:1011342220621

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1011342220621

Navigation