Skip to main content

Advertisement

Log in

Ring-substituted analogs of 3,3′-diindolylmethane (DIM) induce apoptosis and necrosis in androgen-dependent and –independent prostate cancer cells

  • PRECLINICAL STUDIES
  • Published:
Investigational New Drugs Aims and scope Submit manuscript

Summary

We recently reported that novel ring-substituted analogs of 3,3′-diindolylmethane (ring-DIMs) have anti-androgenic and growth inhibitory effects in androgen-dependent prostate cancer cells. The objectives of this study were to confirm the ability of 4,4′- and 7,7′-dibromo- and dichloro-substituted ring-DIMs to inhibit androgen-stimulated proliferation of androgen-dependent LNCaP human prostate cancer cells using a non-invasive, real-time monitoring technique. In addition, their ability to induce apoptotic and necrotic cell death in androgen-dependent as well as -independent (PC-3) prostate cancer cells was studied. Prostate cancer cells were treated with increasing concentrations of DIM and ring-DIMs (0.3–30 μM) and effects on cell proliferation were measured in real-time using an xCELLigence cellular analysis system. Chromatin condensation and loss of membrane integrity were determined by Hoechst and propidium iodide staining, respectively. Apoptotic protein markers were measured by immunoblotting and activation of caspases determined using selective fluorogenic substrates. Intra- and extracellular concentrations of DIM and ring-DIMs were assessed by electrospray ionization tandem mass spectrometry. Ring-DIMs inhibited androgen-stimulated LNCaP cell proliferation and induced apoptosis and necrosis in LNCaP and PC-3 cells with 2–4 fold greater potencies than DIM. DIM and the ring-DIMs increased caspases −3, −8 and −9 activity, elevated expression of Fas, FasL, DR4 and DR5 protein, and induced PARP cleavage in both cell lines. The cytotoxicity of the most potent ring-DIM, 4,4′-dibromoDIM, but not the other compounds was decreased by an inhibitor of caspase −3. The 4,4′-dibromoDIM was primarily found in the extracellular medium, whereas all other compounds were present to a much larger extent in the cell. In conclusion, ring-DIMs inhibited prostate cancer cell growth and induced cell death in LNCaP and PC-3 cells with greater potencies than DIM; they also structure-dependently activated different cell death pathways suggesting that these compounds have clinical potential as chemopreventive and chemotherapeutic agents in prostate cancer, regardless of hormone-dependency.

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
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Siegel R, Naishadham D, Jemal A (2012) Cancer statistics, 2012. CA Cancer J Clin 62:10–29

    Article  PubMed  Google Scholar 

  2. Wysowski DK, Freiman JP, Tourtelot JB, Horton ML 3rd (1993) Fatal and nonfatal hepatotoxicity associated with flutamide. Ann Intern Med 118:860–864

    Article  CAS  PubMed  Google Scholar 

  3. McLeod DG (1997) Tolerability of Nonsteroidal Antiandrogens in the Treatment of Advanced Prostate Cancer. Oncologist 2:18–27

    CAS  PubMed  Google Scholar 

  4. Cohen JH, Kristal AR, Stanford JL (2000) Fruit and vegetable intakes and prostate cancer risk. Journal of the National Cancer Institute 92:61–68

    Article  CAS  PubMed  Google Scholar 

  5. Kristal AR, Lampe JW (2002) Brassica vegetables and prostate cancer risk: a review of the epidemiological evidence. Nutr Cancer 42:1–9

    Article  PubMed  Google Scholar 

  6. Bjeldanes LF, Kim J-Y, Grose KR, Bartholomew JC, Bradfield CA (1991) Aromatic hydrocarbon responsiveness-receptor agonists generated from indole-3-carbinol in vitro and in vivo: comparisons with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Proc Natl Acad Sci USA 88:9543–9547

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  7. De Kruif CA, Marsman JW, Venekamp JC, Falke HE, Noordhoek J, Blaauboer BJ, Wortelboer HM (1991) Structure elucidation of acid reaction products of indole-3-carbinol: detection in vivo and enzyme induction in vitro. Chem-Biol Interact 80:303–315

    Article  PubMed  Google Scholar 

  8. Ge X, Yannai S, Rennert G, Gruener N, Fares FA (1996) 3,3′-Diindolylmethane induces apoptosis in human cancer cells. Biochem Biophys Res Commun 228:153–158

    Article  CAS  PubMed  Google Scholar 

  9. Nachshon-Kedmi M, Fares FA, Yannai S (2004) Therapeutic activity of 3,3′-diindolylmethane on prostate cancer in an in vivo model. Prostate 61:153–160

    Article  CAS  PubMed  Google Scholar 

  10. Heath EI, Heilbrun LK, Li J, Vaishampayan U, Harper F, Pemberton P, Sarkar FH (2010) A phase I dose-escalation study of oral BR-DIM (BioResponse 3,3′- Diindolylmethane) in castrate-resistant, non-metastatic prostate cancer. American journal of translational research 2:402–411

    PubMed Central  CAS  PubMed  Google Scholar 

  11. Le HT, Schaldach CM, Firestone GL, Bjeldanes LF (2003) Plant-derived 3,3′-diindolylmethane is a strong androgen antagonist in human prostate cancer cells. J Biol Chem 278:21136–21145

    Article  CAS  PubMed  Google Scholar 

  12. Li Y, Kong D, Wang Z, Ahmad A, Bao B, Padhye S, Sarkar FH (2011) Inactivation of AR/TMPRSS2-ERG/Wnt signaling networks attenuates the aggressive behavior of prostate cancer cells. Cancer prevention research 4:1495–1506

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  13. Wang TT, Schoene NW, Milner JA, Kim YS (2012) 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 51:244–256

    Article  PubMed  Google Scholar 

  14. Bhuiyan MM, Li Y, Banerjee S, Ahmed F, Wang Z, Ali S, Sarkar FH (2006) 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 66:10064–10072

    Article  CAS  PubMed  Google Scholar 

  15. Chinnakannu K, Chen D, Li Y, Wang Z, Dou QP, Reddy GP, Sarkar FH (2009) Cell cycle-dependent effects of 3,3′-diindolylmethane on proliferation and apoptosis of prostate cancer cells. J Cell Physiol 219:94–99

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  16. Garikapaty VP, Ashok BT, Tadi K, Mittelman A, Tiwari RK (2006) 3,3′-Diindolylmethane downregulates pro-survival pathway in hormone independent prostate cancer. Biochem Biophys Res Commun 340:718–725

    Article  CAS  PubMed  Google Scholar 

  17. Kong D, Banerjee S, Huang W, Li Y, Wang Z, Kim HR, Sarkar FH (2008) Mammalian target of rapamycin repression by 3,3′-diindolylmethane inhibits invasion and angiogenesis in platelet-derived growth factor-D-overexpressing PC3 cells. Cancer Res 68:1927–1934

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  18. Li Y, Wang Z, Kong D, Murthy S, Dou QP, Sheng S, Reddy GP, Sarkar FH (2007) Regulation of FOXO3a/beta-catenin/GSK-3beta signaling by 3,3′-diindolylmethane contributes to inhibition of cell proliferation and induction of apoptosis in prostate cancer cells. J Biol Chem 282:21542–21550

    Article  CAS  PubMed  Google Scholar 

  19. Abdelrahim M, Newman K, Vanderlaag K, Samudio I, Safe S (2006) 3,3′-diindolylmethane (DIM) and its derivatives induce apoptosis in pancreatic cancer cells through endoplasmic reticulum stress-dependent upregulation of DR5. Carcinogenesis 27:717–728

    Article  CAS  PubMed  Google Scholar 

  20. Savino JA 3rd, Evans JF, Rabinowitz D, Auborn KJ, Carter TH (2006) Multiple, disparate roles for calcium signaling in apoptosis of human prostate and cervical cancer cells exposed to diindolylmethane. Mol Cancer Ther 5:556–563

    Article  CAS  PubMed  Google Scholar 

  21. Sun S, Han J, Ralph WM Jr, Chandrasekaran A, Liu K, Auborn KJ, Carter TH (2004) Endoplasmic reticulum stress as a correlate of cytotoxicity in human tumor cells exposed to diindolylmethane in vitro. Cell stress & chaperones 9:76–87

    Article  CAS  Google Scholar 

  22. Cho HJ, Park SY, Kim EJ, Kim JK, Park JH (2011) 3,3′-Diindolylmethane inhibits prostate cancer development in the transgenic adenocarcinoma mouse prostate model. Mol Carcinog 50:100–112

    Article  CAS  PubMed  Google Scholar 

  23. Nachshon-Kedmi M, Yannai S, Haj A, Fares FA (2003) Indole-3-carbinol and 3,3′-diindolylmethane induce apoptosis in human prostate cancer cells. Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association 41:745–752

    Article  CAS  Google Scholar 

  24. Nachshon-Kedmi M, Yannai S, Fares FA (2004) Induction of apoptosis in human prostate cancer cell line, PC3, by 3,3′-diindolylmethane through the mitochondrial pathway. Br J Cancer 91:1358–1363

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  25. Chintharlapalli S, Papineni S, Safe S (2006) 1,1-Bis(3′-indolyl)-1-(p-substituted phenyl)methanes inhibit colon cancer cell and tumor growth through PPARgamma-dependent and PPARgamma-independent pathways. Mol Cancer Ther 5:1362–1370

    Article  CAS  PubMed  Google Scholar 

  26. Cho SD, Yoon K, Chintharlapalli S, Abdelrahim M, Lei P, Hamilton S, Khan S, Ramaiah SK, Safe S (2007) Nur77 agonists induce proapoptotic genes and responses in colon cancer cells through nuclear receptor-dependent and nuclear receptor-independent pathways. Cancer Res 67:674–683

    Article  CAS  PubMed  Google Scholar 

  27. Lei P, Abdelrahim M, Cho SD, Liu S, Chintharlapalli S, Safe S (2008) 1,1-Bis(3′-indolyl)-1-(p-substituted phenyl)methanes inhibit colon cancer cell and tumor growth through activation of c-jun N-terminal kinase. Carcinogenesis 29:1139–1147

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  28. Lei P, Abdelrahim M, Safe S (2006) 1,1-Bis(3′-indolyl)-1-(p-substituted phenyl)methanes inhibit ovarian cancer cell growth through peroxisome proliferator-activated receptor-dependent and independent pathways. Mol Cancer Ther 5:2324–2336

    Article  CAS  PubMed  Google Scholar 

  29. Qin C, Morrow D, Stewart J, Spencer K, Porter W, Smith R 3rd, Phillips T, Abdelrahim M, Samudio I, Safe S (2004) A new class of peroxisome proliferator-activated receptor gamma (PPARgamma) agonists that inhibit growth of breast cancer cells: 1,1-Bis(3′-indolyl)-1-(p-substituted phenyl)methanes. Mol Cancer Ther 3:247–260

    Article  CAS  PubMed  Google Scholar 

  30. Yoon K, Lee SO, Cho SD, Kim K, Khan S, Safe S (2011) Activation of nuclear TR3 (NR4A1) by a diindolylmethane analog induces apoptosis and proapoptotic genes in pancreatic cancer cells and tumors. Carcinogenesis 32:836–842

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  31. Abdelbaqi K, Lack N, Guns ET, Kotha L, Safe S, Sanderson JT (2011) Antiandrogenic and growth inhibitory effects of ring-substituted analogs of 3,3′-diindolylmethane (Ring-DIMs) in hormone-responsive LNCaP human prostate cancer cells. Prostate 71:1401–1412

    Article  CAS  PubMed  Google Scholar 

  32. Bourque SD, Titorenko VI (2009) A quantitative assessment of the yeast lipidome using electrospray ionization mass spectrometry. Journal of visualized experiments, JoVE

    Google Scholar 

  33. Bratton SB, MacFarlane M, Cain K, Cohen GM (2000) Protein complexes activate distinct caspase cascades in death receptor and stress-induced apoptosis. Exp Cell Res 256:27–33

    Article  CAS  PubMed  Google Scholar 

  34. Lavrik IN, Krammer PH (2012) Regulation of CD95/Fas signaling at the DISC. Cell death and differentiation 19:36–41

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  35. Rath PC, Aggarwal BB (1999) TNF-induced signaling in apoptosis. J Clin Immunol 19:350–364

    Article  CAS  PubMed  Google Scholar 

  36. Jiang X, Wang X (2004) Cytochrome C-mediated apoptosis. Annu Rev Biochem 73:87–106

    Article  CAS  PubMed  Google Scholar 

  37. Jourdain A, Martinou JC (2009) Mitochondrial outer-membrane permeabilization and remodelling in apoptosis. Int J Biochem Cell Biol 41:1884–1889

    Article  CAS  PubMed  Google Scholar 

  38. Ow YP, Green DR, Hao Z, Mak TW (2008) Cytochrome c: functions beyond respiration. Nature reviews. Molecular cell biology 9:532–542

    CAS  PubMed  Google Scholar 

  39. Tait SW, Green DR (2010) Mitochondria and cell death: outer membrane permeabilization and beyond. Nature reviews. Molecular cell biology 11:621–632

    CAS  PubMed  Google Scholar 

  40. Cohen GM (1997) Caspases: the executioners of apoptosis. Biochem J 326(Pt 1):1–16

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  41. Slee EA, Harte MT, Kluck RM, Wolf BB, Casiano CA, Newmeyer DD, Wang HG, Reed JC, Nicholson DW, Alnemri ES, Green DR, Martin SJ (1999) Ordering the cytochrome c-initiated caspase cascade: hierarchical activation of caspases-2, -3, -6, -7, -8, and -10 in a caspase-9-dependent manner. J Cell Biol 144:281–292

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  42. Liu H, Dong A, Gao C, Tan C, Xie Z, Zu X, Qu L, Jiang Y (2010) New synthetic flavone derivatives induce apoptosis of hepatocarcinoma cells. Bioorg Med Chem 18:6322–6328

    Article  CAS  PubMed  Google Scholar 

  43. Yan YQ, Xu QZ, Wang L, Sui JL, Bai B, Zhou PK (2006) Vanillin derivative 6-bromine-5-hydroxy-4-methoxybenzaldehyde-elicited apoptosis and G2/M arrest of Jurkat cells proceeds concurrently with DNA-PKcs cleavage and Akt inactivation. Int J Oncol 29:1167–1172

    CAS  PubMed  Google Scholar 

  44. Cande C, Cohen I, Daugas E, Ravagnan L, Larochette N, Zamzami N, Kroemer G (2002) Apoptosis-inducing factor (AIF): a novel caspase-independent death effector released from mitochondria. Biochimie 84:215–222

    Article  CAS  PubMed  Google Scholar 

  45. Kim R, Emi M, Tanabe K (2005) Caspase-dependent and -independent cell death pathways after DNA damage (Review). Oncology reports 14:595–599

    CAS  PubMed  Google Scholar 

  46. Li LY, Luo X, Wang X (2001) Endonuclease G is an apoptotic DNase when released from mitochondria. Nature 412:95–99

    Article  CAS  PubMed  Google Scholar 

  47. Lorenzo HK, Susin SA (2004) Mitochondrial effectors in caspase-independent cell death. FEBS Lett 557:14–20

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was made possible by an operating grant from the Canadian Institutes of Health Research (CIHR grant no. MOP-115019). The authors declare to have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to J. T. Sanderson.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Goldberg, A.A., Titorenko, V.I., Beach, A. et al. Ring-substituted analogs of 3,3′-diindolylmethane (DIM) induce apoptosis and necrosis in androgen-dependent and –independent prostate cancer cells. Invest New Drugs 32, 25–36 (2014). https://doi.org/10.1007/s10637-013-9979-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10637-013-9979-y

Keywords

Navigation