Skip to main content

Advertisement

Log in

Phytochemicals from Cruciferous Vegetables, Epigenetics, and Prostate Cancer Prevention

  • Review Article
  • Theme: Natural Products Drug Discovery in Cancer Prevention
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

Epidemiological evidence has demonstrated a reduced risk of prostate cancer associated with cruciferous vegetable intake. Follow-up studies have attributed this protective activity to the metabolic products of glucosinolates, a class of secondary metabolites produced by crucifers. The metabolic products of glucoraphanin and glucobrassicin, sulforaphane, and indole-3-carbinol respectively, have been the subject of intense investigation by cancer researchers. Sulforaphane and indole-3-carbinol inhibit prostate cancer by both blocking initiation and suppressing prostate cancer progression in vitro and in vivo. Research has largely focused on the anti-initiation and cytoprotective effects of sulforaphane and indole-3-carbinol through induction of phases I and II detoxification pathways. With regards to suppressive activity, research has focused on the ability of sulforaphane and indole-3-carbinol to antagonize cell signaling pathways known to be dysregulated in prostate cancer. Recent investigations have characterized the ability of sulforaphane and indole-3-carbinol derivatives to modulate the activity of enzymes controlling the epigenetic status of prostate cancer cells. In this review, we will summarize the well-established, “classic” non-epigenetic targets of sulforaphane and indole-3-carbinol, and highlight more recent evidence supporting these phytochemicals as epigenetic modulators for prostate cancer chemoprevention.

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.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

REFERENCES

  1. Richman EL, Carroll PR, Chan JM. Vegetable and fruit intake after diagnosis and risk of prostate cancer progression. Int J Cancer. 2011;131(1):201–10.

    Article  PubMed  Google Scholar 

  2. Liu B, Mao Q, Cao M, Xie L. Cruciferous vegetables intake and risk of prostate cancer: a meta-analysis. Int J Urol. 2012;19(2):134–41.

    Article  PubMed  Google Scholar 

  3. Steinbrecher A, Nimptsch K, Husing A, Rohrmann S, Linseisen J. Dietary glucosinolate intake and risk of prostate cancer in the EPIC-Heidelberg cohort study. Int J Cancer. 2009;125(9):2179–86.

    Article  PubMed  CAS  Google Scholar 

  4. Howlader N, Noone AM, Krapcho M, Neyman N, Aminou R, Altekruse SF, et al., editors. SEER Cancer Statistics Review, 1975–2009 (Vintage 2009 Populations). 2012.

  5. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. GLOBOCAN 2008 v2.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 10 (Internet). International Agency for Research on Cancer. 2010. Available from: http://globocan.iarc.fr. Accessed 12/11/2012

  6. Shimizu H, Ross RK, Bernstein L, Yatani R, Henderson BE, Mack TM. Cancers of the prostate and breast among Japanese and white immigrants in Los Angeles County. Br J Cancer. 1991;63(6):963–6.

    Article  PubMed  CAS  Google Scholar 

  7. Lee J, Demissie K, Lu SE, Rhoads GG. Cancer incidence among Korean-American immigrants in the United States and native Koreans in South Korea. Cancer Control. 2007;14(1):78–85.

    PubMed  Google Scholar 

  8. IARC. IARC: cruciferous vegetables, isothiocyanates and indoles, vol. 9. Lyon: IARC; 2004.

    Google Scholar 

  9. McNaughton SA, Marks GC. Development of a food composition database for the estimation of dietary intakes of glucosinolates, the biologically active constituents of cruciferous vegetables. Br J Nutr. 2003;90(3):687–97.

    Article  PubMed  CAS  Google Scholar 

  10. Hanlon N, Coldham N, Gielbert A, Sauer MJ, Ioannides C. Repeated intake of broccoli does not lead to higher plasma levels of sulforaphane in human volunteers. Cancer Lett. 2009;284(1):15–20.

    Article  PubMed  CAS  Google Scholar 

  11. Hauder J, Winkler S, Bub A, Rufer CE, Pignitter M, Somoza V. LC-MS/MS quantification of sulforaphane and indole-3-carbinol metabolites in human plasma and urine after dietary intake of selenium-fortified broccoli. J Agric Food Chem. 2011;59(15):8047–57.

    Article  PubMed  CAS  Google Scholar 

  12. Veeranki OL, Bhattacharya A, Marshall JR, Zhang Y. Organ-specific exposure and response to sulforaphane, a key chemopreventive ingredient in broccoli: implications for cancer prevention. Br J Nutr. 2012;2:1–8.

    Google Scholar 

  13. Clarke JD, Hsu A, Williams DE, Dashwood RH, Stevens JF, Yamamoto M, et al. Metabolism and tissue distribution of sulforaphane in Nrf2 knockout and wild-type mice. Pharm Res. 2011;28(12):3171–9.

    Article  PubMed  CAS  Google Scholar 

  14. Keum YS, Khor TO, Lin W, Shen G, Kwon KH, Barve A, et al. Pharmacokinetics and pharmacodynamics of broccoli sprouts on the suppression of prostate cancer in transgenic adenocarcinoma of mouse prostate (TRAMP) mice: implication of induction of Nrf2, HO-1 and apoptosis and the suppression of Akt-dependent kinase pathway. Pharm Res. 2009;26(10):2324–31.

    Article  PubMed  CAS  Google Scholar 

  15. Traka MH, Spinks CA, Doleman JF, Melchini A, Ball RY, Mills RD, et al. The dietary isothiocyanate sulforaphane modulates gene expression and alternative gene splicing in a PTEN null preclinical murine model of prostate cancer. Mol Cancer. 2010;9:189.

    Article  PubMed  Google Scholar 

  16. Traka M, Gasper AV, Melchini A, Bacon JR, Needs PW, Frost V, et al. Broccoli consumption interacts with GSTM1 to perturb oncogenic signalling pathways in the prostate. PLoS One. 2008;3(7):e2568.

    Article  PubMed  Google Scholar 

  17. Grose KR, Bjeldanes LF. Oligomerization of indole-3-carbinol in aqueous acid. Chem Res Toxicol. 1992;5(2):188–93.

    Article  PubMed  CAS  Google Scholar 

  18. Anderton MJ, Manson MM, Verschoyle RD, Gescher A, Lamb JH, Farmer PB, et al. Pharmacokinetics and tissue disposition of indole-3-carbinol and its acid condensation products after oral administration to mice. Clin Cancer Res. 2004;10(15):5233–41.

    Article  PubMed  CAS  Google Scholar 

  19. Reed GA, Arneson DW, Putnam WC, Smith HJ, Gray JC, Sullivan DK, et al. Single-dose and multiple-dose administration of indole-3-carbinol to women: pharmacokinetics based on 3,3′-diindolylmethane. Cancer Epidemiol Biomarkers Prev. 2006;15(12):2477–81.

    Article  PubMed  CAS  Google Scholar 

  20. Souli E, Machluf M, Morgenstern A, Sabo E, Yannai S. Indole-3-carbinol (I3C) exhibits inhibitory and preventive effects on prostate tumors in mice. Food Chem Toxicol. 2008;46(3):863–70.

    Article  PubMed  CAS  Google Scholar 

  21. Bradlow HL, Zeligs MA. Diindolylmethane (DIM) spontaneously forms from indole-3-carbinol (I3C) during cell culture experiments. In Vivo (Athens, Greece). 2010;24(4):387–91.

    CAS  Google Scholar 

  22. Bradlow HL. Review. Indole-3-carbinol as a chemoprotective agent in breast and prostate cancer. In Vivo (Athens, Greece). 2008;22(4):441–5.

    CAS  Google Scholar 

  23. Cho HJ, Park SY, Kim EJ, Kim JK, Park JH. 3,3′-diindolylmethane inhibits prostate cancer development in the transgenic adenocarcinoma mouse prostate model. Mol Carcinog. 2011;50(2):100–12.

    Article  PubMed  CAS  Google Scholar 

  24. Gasper AV, Al-Janobi A, Smith JA, Bacon JR, Fortun P, Atherton C, et al. Glutathione S-transferase M1 polymorphism and metabolism of sulforaphane from standard and high-glucosinolate broccoli. Am J Clin Nutr. 2005;82(6):1283–91.

    PubMed  CAS  Google Scholar 

  25. Heath EI, Heilbrun LK, Li J, Vaishampayan U, Harper F, Pemberton P, et al. A phase I dose-escalation study of oral BR-DIM (BioResponse 3,3′-diindolylmethane) in castrate-resistant, non-metastatic prostate cancer. Am J Trans Res. 2010;2(4):402–11.

    CAS  Google Scholar 

  26. Brooks JD, Paton VG, Vidanes G. Potent induction of phase 2 enzymes in human prostate cells by sulforaphane. Cancer Epidemiol Biomarkers Prev. 2001;10(9):949–54.

    PubMed  CAS  Google Scholar 

  27. Fahey JW, Talalay P. Antioxidant functions of sulforaphane: a potent inducer of phase II detoxication enzymes. Food Chem Toxicol. 1999;37(9-10):973–9.

    Article  PubMed  CAS  Google Scholar 

  28. Guerrero-Beltran CE, Calderon-Oliver M, Pedraza-Chaverri J, Chirino YI. Protective effect of sulforaphane against oxidative stress: recent advances. Exp Toxicol Pathol. 2012;64(5):503–8.

    Article  PubMed  CAS  Google Scholar 

  29. Wang TT, Schoene NW, Milner JA, Kim YS. Broccoli-derived phytochemicals indole-3-carbinol and 3,3′-diindolylmethane exerts concentration-dependent pleiotropic effects on prostate cancer cells: comparison with other cancer preventive phytochemicals. Mol Carcinog. 2012;51(3):244–56.

    Article  PubMed  Google Scholar 

  30. Ernst IM, Schuemann C, Wagner AE, Rimbach G. 3,3′-diindolylmethane but not indole-3-carbinol activates Nrf2 and induces Nrf2 target gene expression in cultured murine fibroblasts. Free Radic Res. 2011;45(8):941–9.

    Article  PubMed  CAS  Google Scholar 

  31. Saw CL, Cintron M, Wu TY, Guo Y, Huang Y, Jeong WS, et al. Pharmacodynamics of dietary phytochemical indoles I3C and DIM: induction of Nrf2-mediated phase II drug metabolizing and antioxidant genes and synergism with isothiocyanates. Biopharm Drug Dispos. 2011;32(5):289–300.

    Article  PubMed  CAS  Google Scholar 

  32. Reed GA, Peterson KS, Smith HJ, Gray JC, Sullivan DK, Mayo MS, et al. A phase I study of indole-3-carbinol in women: tolerability and effects. Cancer Epidemiol Biomarkers Prev. 2005;14(8):1953–60.

    Article  PubMed  CAS  Google Scholar 

  33. Dalessandri KM, Firestone GL, Fitch MD, Bradlow HL, Bjeldanes LF. Pilot study: effect of 3,3′-diindolylmethane supplements on urinary hormone metabolites in postmenopausal women with a history of early-stage breast cancer. Nutr Cancer. 2004;50(2):161–7.

    Article  PubMed  CAS  Google Scholar 

  34. Michnovicz JJ, Adlercreutz H, Bradlow HL. Changes in levels of urinary estrogen metabolites after oral indole-3-carbinol treatment in humans. J Natl Cancer Inst. 1997;89(10):718–23.

    Article  PubMed  CAS  Google Scholar 

  35. Michnovicz JJ, Bradlow HL. Altered estrogen metabolism and excretion in humans following consumption of indole-3-carbinol. Nutr Cancer. 1991;16(1):59–66.

    Article  PubMed  CAS  Google Scholar 

  36. Fowke JH, Longcope C, Hebert JR. Brassica vegetable consumption shifts estrogen metabolism in healthy postmenopausal women. Cancer Epidemiol Biomarkers Prev. 2000;9(8):773–9.

    PubMed  CAS  Google Scholar 

  37. Castagnetta LA, Miceli MD, Sorci CM, Pfeffer U, Farruggio R, Oliveri G, et al. Growth of LNCaP human prostate cancer cells is stimulated by estradiol via its own receptor. Endocrinology. 1995;136(5):2309–19.

    Article  PubMed  CAS  Google Scholar 

  38. Dubrovska A, Kim S, Salamone RJ, Walker JR, Maira SM, Garcia-Echeverria C, et al. The role of PTEN/Akt/PI3K signaling in the maintenance and viability of prostate cancer stem-like cell populations. Proc Natl Acad Sci U S A. 2009;106(1):268–73.

    Article  PubMed  CAS  Google Scholar 

  39. Majumder PK, Sellers WR. Akt-regulated pathways in prostate cancer. Oncogene. 2005;24(50):7465–74.

    Article  PubMed  CAS  Google Scholar 

  40. Sarker D, Reid AH, Yap TA, de Bono JS. Targeting the PI3K/AKT pathway for the treatment of prostate cancer. Clin Cancer Res. 2009;15(15):4799–805.

    Article  PubMed  CAS  Google Scholar 

  41. Kinkade CW, Castillo-Martin M, Puzio-Kuter A, Yan J, Foster TH, Gao H, et al. Targeting AKT/mTOR and ERK MAPK signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model. J Clin Investig. 2008;118(9):3051–64.

    PubMed  CAS  Google Scholar 

  42. Morgan TM, Koreckij TD, Corey E. Targeted therapy for advanced prostate cancer: inhibition of the PI3K/Akt/mTOR pathway. Curr Cancer Drug Targets. 2009;9(2):237–49.

    Article  PubMed  CAS  Google Scholar 

  43. Wiczk A, Hofman D, Konopa G, Herman-Antosiewicz A. Sulforaphane, a cruciferous vegetable-derived isothiocyanate, inhibits protein synthesis in human prostate cancer cells. Biochim Biophys Acta. 2012;1823(8):1295–305.

    Article  PubMed  CAS  Google Scholar 

  44. Martelli AM, Tabellini G, Bressanin D, Ognibene A, Goto K, Cocco L, et al. The emerging multiple roles of nuclear Akt. Biochim Biophys Acta. 2012;1823(12):2168–78.

    Article  PubMed  CAS  Google Scholar 

  45. Song G, Ouyang G, Bao S. The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med. 2005;9(1):59–71.

    Article  PubMed  CAS  Google Scholar 

  46. Xu C, Shen G, Chen C, Gelinas C, Kong AN. Suppression of NF-kappaB and NF-kappaB-regulated gene expression by sulforaphane and PEITC through IkappaBalpha, IKK pathway in human prostate cancer PC-3 cells. Oncogene. 2005;24(28):4486–95.

    Article  PubMed  CAS  Google Scholar 

  47. Kennedy SG, Wagner AJ, Conzen SD, Jordan J, Bellacosa A, Tsichlis PN, et al. The PI 3-kinase/Akt signaling pathway delivers an anti-apoptotic signal. Genes Dev. 1997;11(6):701–13.

    Article  PubMed  CAS  Google Scholar 

  48. Xu C, Shen G, Yuan X, Kim JH, Gopalkrishnan A, Keum YS, et al. ERK and JNK signaling pathways are involved in the regulation of activator protein 1 and cell death elicited by three isothiocyanates in human prostate cancer PC-3 cells. Carcinogenesis. 2006;27(3):437–45.

    Article  PubMed  Google Scholar 

  49. Choi S, Lew KL, Xiao H, Herman-Antosiewicz A, Xiao D, Brown CK, et al. d,l-sulforaphane-induced cell death in human prostate cancer cells is regulated by inhibitor of apoptosis family proteins and Apaf-1. Carcinogenesis. 2007;28(1):151–62.

    Article  PubMed  CAS  Google Scholar 

  50. Dan HC, Cooper MJ, Cogswell PC, Duncan JA, Ting JP, Baldwin AS. Akt-dependent regulation of NF-{kappa}B is controlled by mTOR and Raptor in association with IKK. Genes Dev. 2008;22(11):1490–500.

    Article  PubMed  CAS  Google Scholar 

  51. Choi S, Singh SV. Bax and Bak are required for apoptosis induction by sulforaphane, a cruciferous vegetable-derived cancer chemopreventive agent. Cancer Res. 2005;65(5):2035–43.

    Article  PubMed  CAS  Google Scholar 

  52. Chu ZL, McKinsey TA, Liu L, Gentry JJ, Malim MH, Ballard DW. Suppression of tumor necrosis factor-induced cell death by inhibitor of apoptosis c-IAP2 is under NF-kappaB control. Proc Natl Acad Sci U S A. 1997;94(19):10057–62.

    Article  PubMed  CAS  Google Scholar 

  53. Stehlik C, de Martin R, Kumabashiri I, Schmid JA, Binder BR, Lipp J. Nuclear factor (NF)-kappaB-regulated X-chromosome-linked iap gene expression protects endothelial cells from tumor necrosis factor alpha-induced apoptosis. J Exp Med. 1998;188(1):211–6.

    Article  PubMed  CAS  Google Scholar 

  54. McEleny K, Coffey R, Morrissey C, Williamson K, Zangemeister-Wittke U, Fitzpatrick JM, et al. An antisense oligonucleotide to cIAP-1 sensitizes prostate cancer cells to fas and TNFalpha mediated apoptosis. Prostate. 2004;59(4):419–25.

    Article  PubMed  CAS  Google Scholar 

  55. Xiao D, Powolny AA, Antosiewicz J, Hahm ER, Bommareddy A, Zeng Y, et al. Cellular responses to cancer chemopreventive agent d,l-sulforaphane in human prostate cancer cells are initiated by mitochondrial reactive oxygen species. Pharm Res. 2009;26(7):1729–38.

    Article  PubMed  CAS  Google Scholar 

  56. Singh AV, Xiao D, Lew KL, Dhir R, Singh SV. Sulforaphane induces caspase-mediated apoptosis in cultured PC-3 human prostate cancer cells and retards growth of PC-3 xenografts in vivo. Carcinogenesis. 2004;25(1):83–90.

    Article  PubMed  CAS  Google Scholar 

  57. Singh SV, Warin R, Xiao D, Powolny AA, Stan SD, Arlotti JA, et al. Sulforaphane inhibits prostate carcinogenesis and pulmonary metastasis in TRAMP mice in association with increased cytotoxicity of natural killer cells. Cancer Res. 2009;69(5):2117–25.

    Article  PubMed  CAS  Google Scholar 

  58. Clarke JD, Dashwood RH, Ho E. Multi-targeted prevention of cancer by sulforaphane. Cancer Lett. 2008;269(2):291–304.

    Article  PubMed  CAS  Google Scholar 

  59. Juge N, Mithen RF, Traka M. Molecular basis for chemoprevention by sulforaphane: a comprehensive review. Cell Mol Life Sci. 2007;64(9):1105–27.

    Article  PubMed  CAS  Google Scholar 

  60. Chinni SR, Li Y, Upadhyay S, Koppolu PK, Sarkar FH. Indole-3-carbinol (I3C) induced cell growth inhibition, G1 cell cycle arrest and apoptosis in prostate cancer cells. Oncogene. 2001;20(23):2927–36.

    Article  PubMed  CAS  Google Scholar 

  61. Nachshon-Kedmi M, Yannai S, Haj A, Fares FA. Indole-3-carbinol and 3,3′-diindolylmethane induce apoptosis in human prostate cancer cells. Food Chem Toxicol. 2003;41(6):745–52.

    Article  PubMed  CAS  Google Scholar 

  62. Vivar OI, Lin CL, Firestone GL, Bjeldanes LF. 3,3′-diindolylmethane induces a G(1) arrest in human prostate cancer cells irrespective of androgen receptor and p53 status. Biochem Pharmacol. 2009;78(5):469–76.

    Article  PubMed  CAS  Google Scholar 

  63. Nachshon-Kedmi M, Yannai S, Fares FA. Induction of apoptosis in human prostate cancer cell line, PC3, by 3,3′-diindolylmethane through the mitochondrial pathway. Br J Cancer. 2004;91(7):1358–63.

    Article  PubMed  CAS  Google Scholar 

  64. Garikapaty VP, Ashok BT, Tadi K, Mittelman A, Tiwari RK. 3,3′-diindolylmethane downregulates pro-survival pathway in hormone independent prostate cancer. Biochem Biophys Res Commun. 2006;340(2):718–25.

    Article  PubMed  CAS  Google Scholar 

  65. Bhuiyan MM, Li Y, Banerjee S, Ahmed F, Wang Z, Ali S, et al. Down-regulation of androgen receptor by 3,3′-diindolylmethane contributes to inhibition of cell proliferation and induction of apoptosis in both hormone-sensitive LNCaP and insensitive C4-2B prostate cancer cells. Cancer Res. 2006;66(20):10064–72.

    Article  PubMed  CAS  Google Scholar 

  66. Kong D, Banerjee S, Huang W, Li Y, Wang Z, Kim HR, et al. Mammalian target of rapamycin repression by 3,3′-diindolylmethane inhibits invasion and angiogenesis in platelet-derived growth factor-d-overexpressing PC3 cells. Cancer Res. 2008;68(6):1927–34.

    Article  PubMed  CAS  Google Scholar 

  67. Kong D, Li Y, Wang Z, Banerjee S, Sarkar FH. Inhibition of angiogenesis and invasion by 3,3′-diindolylmethane is mediated by the nuclear factor-kappaB downstream target genes MMP-9 and uPA that regulated bioavailability of vascular endothelial growth factor in prostate cancer. Cancer Res. 2007;67(7):3310–9.

    Article  PubMed  CAS  Google Scholar 

  68. Li Y, Chinni SR, Sarkar FH. Selective growth regulatory and pro-apoptotic effects of DIM is mediated by AKT and NF-kappaB pathways in prostate cancer cells. Front Biosci. 2005;10:236–43.

    Article  PubMed  CAS  Google Scholar 

  69. Chen D, Banerjee S, Cui QC, Kong D, Sarkar FH, Dou QP. Activation of AMP-activated protein kinase by 3,3′-diindolylmethane (DIM) is associated with human prostate cancer cell death in vitro and in vivo. PLoS One. 2012;7(10):e47186.

    Article  PubMed  CAS  Google Scholar 

  70. Le HT, Schaldach CM, Firestone GL, Bjeldanes LF. Plant-derived 3,3′-diindolylmethane is a strong androgen antagonist in human prostate cancer cells. J Biol Chem. 2003;278(23):21136–45.

    Article  PubMed  CAS  Google Scholar 

  71. Baylin SB, Jones PA. A decade of exploring the cancer epigenome—biological and translational implications. Nat Rev. 2011;11(10):726–34.

    Article  CAS  Google Scholar 

  72. Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31(1):27–36.

    Article  PubMed  CAS  Google Scholar 

  73. Kim HJ, Bae SC. Histone deacetylase inhibitors: molecular mechanisms of action and clinical trials as anti-cancer drugs. Am J Transl Res. 2011;3(2):166–79.

    PubMed  CAS  Google Scholar 

  74. Dashwood RH, Myzak MC, Ho E. Dietary HDAC inhibitors: time to rethink weak ligands in cancer chemoprevention? Carcinogenesis. 2006;27(2):344–9.

    Article  PubMed  CAS  Google Scholar 

  75. Weichert W, Roske A, Gekeler V, Beckers T, Stephan C, Jung K, et al. Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer. 2008;98(3):604–10.

    Article  PubMed  CAS  Google Scholar 

  76. Gibbs A, Schwartzman J, Deng V, Alumkal J. Sulforaphane destabilizes the androgen receptor in prostate cancer cells by inactivating histone deacetylase 6. Proc Natl Acad Sci U S A. 2009;106(39):16663–8.

    Article  PubMed  CAS  Google Scholar 

  77. Noonan EJ, Place RF, Pookot D, Basak S, Whitson JM, Hirata H, et al. miR-449a targets HDAC-1 and induces growth arrest in prostate cancer. Oncogene. 2009;28(14):1714–24.

    Article  PubMed  CAS  Google Scholar 

  78. Myzak MC, Hardin K, Wang R, Dashwood RH, Ho E. Sulforaphane inhibits histone deacetylase activity in BPH-1, LnCaP and PC-3 prostate epithelial cells. Carcinogenesis. 2006;27(4):811–9.

    Article  PubMed  CAS  Google Scholar 

  79. Myzak MC, Tong P, Dashwood WM, Dashwood RH, Ho E. Sulforaphane retards the growth of human PC-3 xenografts and inhibits HDAC activity in human subjects. Exp Biol Med (Maywood, NJ). 2007;232(2):227–34.

    CAS  Google Scholar 

  80. Clarke JD, Hsu A, Yu Z, Dashwood RH, Ho E. Differential effects of sulforaphane on histone deacetylases, cell cycle arrest and apoptosis in normal prostate cells versus hyperplastic and cancerous prostate cells. Mol Nutr Food Res. 2011;55(7):999–1009.

    Article  PubMed  CAS  Google Scholar 

  81. Hsu A, Wong CP, Yu Z, Williams DE, Dashwood RH, Ho E. Promoter de-methylation of cyclin D2 by sulforaphane in prostate cancer cells. Clin Epigenetics. 2011;3:3.

    Article  PubMed  CAS  Google Scholar 

  82. Padar A, Sathyanarayana UG, Suzuki M, Maruyama R, Hsieh JT, Frenkel EP, et al. Inactivation of cyclin D2 gene in prostate cancers by aberrant promoter methylation. Clin Cancer Res. 2003;9(13):4730–4.

    PubMed  CAS  Google Scholar 

  83. Meeran SM, Patel SN, Tollefsbol TO. Sulforaphane causes epigenetic repression of hTERT expression in human breast cancer cell lines. PLoS One. 2010;5(7):e11457.

    Article  PubMed  Google Scholar 

  84. Valenzuela-Fernandez A, Cabrero JR, Serrador JM, Sanchez-Madrid F. HDAC6: a key regulator of cytoskeleton, cell migration and cell-cell interactions. Trends Cell Biol. 2008;18(6):291–7.

    Article  PubMed  CAS  Google Scholar 

  85. Azarenko O, Okouneva T, Singletary KW, Jordan MA, Wilson L. Suppression of microtubule dynamic instability and turnover in MCF7 breast cancer cells by sulforaphane. Carcinogenesis. 2008;29(12):2360–8.

    Article  PubMed  CAS  Google Scholar 

  86. Mi L, Xiao Z, Hood BL, Dakshanamurthy S, Wang X, Govind S, et al. Covalent binding to tubulin by isothiocyanates. A mechanism of cell growth arrest and apoptosis. J Biol Chem. 2008;283(32):22136–46.

    Article  PubMed  CAS  Google Scholar 

  87. Xiao Z, Mi L, Chung FL, Veenstra TD. Proteomic analysis of covalent modifications of tubulins by isothiocyanates. J Nutr. 2012;142(7):1377S–81S.

    Article  PubMed  CAS  Google Scholar 

  88. Shankar S, Ganapathy S, Srivastava RK. Sulforaphane enhances the therapeutic potential of TRAIL in prostate cancer orthotopic model through regulation of apoptosis, metastasis, and angiogenesis. Clin Cancer Res. 2008;14(21):6855–66.

    Article  PubMed  CAS  Google Scholar 

  89. Ai J, Wang Y, Dar JA, Liu J, Liu L, Nelson JB, et al. HDAC6 regulates androgen receptor hypersensitivity and nuclear localization via modulating Hsp90 acetylation in castration-resistant prostate cancer. Mol Endocrinol (Baltimore, Md). 2009;23(12):1963–72.

    Article  CAS  Google Scholar 

  90. Beaver LM, Yu TW, Sokolowski EI, Williams DE, Dashwood RH, Ho E. 3,3′-diindolylmethane, but not indole-3-carbinol, inhibits histone deacetylase activity in prostate cancer cells. Toxicol Appl Pharmacol. 2012;263(3):345–51.

    Article  PubMed  CAS  Google Scholar 

  91. Li Y, Li X, Guo B. Chemopreventive agent 3,3′-diindolylmethane selectively induces proteasomal degradation of class I histone deacetylases. Cancer Res. 2010;70(2):646–54.

    Article  PubMed  CAS  Google Scholar 

  92. Kong D, Heath E, Chen W, Cher ML, Powell I, Heilbrun L, et al. Loss of let-7 up-regulates EZH2 in prostate cancer consistent with the acquisition of cancer stem cell signatures that are attenuated by BR-DIM. PLoS One. 2012;7(3):e33729.

    Article  PubMed  CAS  Google Scholar 

  93. Bachmann IM, Halvorsen OJ, Collett K, Stefansson IM, Straume O, Haukaas SA, et al. EZH2 expression is associated with high proliferation rate and aggressive tumor subgroups in cutaneous melanoma and cancers of the endometrium, prostate, and breast. J Clin Oncol. 2006;24(2):268–73.

    Article  PubMed  CAS  Google Scholar 

  94. van Leenders GJ, Dukers D, Hessels D, van den Kieboom SW, Hulsbergen CA, Witjes JA, et al. Polycomb-group oncogenes EZH2, BMI1, and RING1 are overexpressed in prostate cancer with adverse pathologic and clinical features. Eur Urol. 2007;52(2):455–63.

    Article  PubMed  Google Scholar 

  95. Kong D, Heath E, Chen W, Cher M, Powell I, Heilbrun L, et al. Epigenetic silencing of miR-34a in human prostate cancer cells and tumor tissue specimens can be reversed by BR-DIM treatment. Am J Transl Res. 2012;4(1):14–23.

    PubMed  CAS  Google Scholar 

  96. Fimognari C, Hrelia P. Sulforaphane as a promising molecule for fighting cancer. Mutat Res. 2007;635(2-3):90–104.

    Article  PubMed  CAS  Google Scholar 

  97. Weng JR, Tsai CH, Kulp SK, Chen CS. Indole-3-carbinol as a chemopreventive and anti-cancer agent. Cancer Lett. 2008;262(2):153–63.

    Article  PubMed  CAS  Google Scholar 

  98. Yu S, Khor TO, Cheung KL, Li W, Wu TY, Huang Y, et al. Nrf2 expression is regulated by epigenetic mechanisms in prostate cancer of TRAMP mice. PLoS One. 2010;5(1):e8579.

    Article  PubMed  Google Scholar 

  99. Choudhary C, Kumar C, Gnad F, Nielsen ML, Rehman M, Walther TC, et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science (New York, NY). 2009;325(5942):834–40.

    Article  CAS  Google Scholar 

  100. Bantscheff M, Hopf C, Savitski MM, Dittmann A, Grandi P, Michon AM, et al. Chemoproteomics profiling of HDAC inhibitors reveals selective targeting of HDAC complexes. Nat Biotechnol. 2011;29(3):255–65.

    Article  PubMed  CAS  Google Scholar 

Download references

ACKNOWLEDGMENTS

Our work is funded by NIH grants CA90890, CA65525, CA122906, CA122959, and CA80176 and by National Institute of Environmental Health Sciences (NIEHS) Center grant P30 ES00210.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Emily Ho.

Additional information

Guest Editors: Ah-Ng Tony Kong and Chi Chen

Rights and permissions

Reprints and permissions

About this article

Cite this article

W. Watson, G., M. Beaver, L., E. Williams, D. et al. Phytochemicals from Cruciferous Vegetables, Epigenetics, and Prostate Cancer Prevention. AAPS J 15, 951–961 (2013). https://doi.org/10.1208/s12248-013-9504-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1208/s12248-013-9504-4

KEY WORDS

Navigation