Skip to main content

Advertisement

Log in

Plumbagin-induced Apoptosis in Human Prostate Cancer Cells is Associated with Modulation of Cellular Redox Status and Generation of Reactive Oxygen Species

  • Research Paper
  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose

To investigate the mechanism of human prostate cancer cell growth inhibition by plumbagin, a constituent of the widely used medicinal herb Plumbago zeylanica L.

Materials and Methods

Cell viability was determined by trypan blue dye exclusion assay. Apoptosis induction was assessed by analysis of cytoplasmic histone-associated DNA fragmentation. Cell cycle distribution and generation of reactive oxygen species (ROS) were determined by flow cytometry. The effect of plumbagin treatment on cellular redox status was determined by analysis of intracellular glutathione (GSH) levels and expression of genes involved in ROS metabolism.

Results

Plumbagin treatment decreased viability of human prostate cancer cells (PC-3, LNCaP, and C4-2) irrespective of their androgen responsiveness or p53 status. Plumbagin-mediated decrease in cell viability correlated with apoptosis induction, which was accompanied by ROS generation and depletion of intracellular GSH levels. Pretreatment of cells with the antioxidant N-acetylcysteine inhibited plumbagin-mediated ROS generation and apoptosis. Plumbagin treatment also resulted in altered expression of genes responsible for ROS metabolism, including superoxide dismutase 2 (Mn-SOD).

Conclusion

The present study points towards an important role of ROS in plumbagin-induced apoptosis in human prostate cancer cells.

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

Similar content being viewed by others

Abbreviations

DCF:

2′,7′-dichlorofluorescein

DMSO:

dimethyl sulfoxide

FBS:

fetal bovine serum

GSH:

glutathione

H2DCFDA:

6-carboxy-2′,7′-dichlorodihydrofluorescein diacetate

Mn-SOD:

manganese superoxide dismutase

NAC:

N-acetylcysteine

NF-κB:

nuclear factor-κB

PBS:

phosphate buffered saline

ROS:

reactive oxygen species

References

  1. A. Jemal, R. Siegel, E. Ward, T. Murray, J. Xu, and M. J. Thun. Cancer statistics. CA Cancer J. Clin 57:43–66 (2007).

    PubMed  Google Scholar 

  2. W. G. Nelson, A. M. De Marzo, and W. B. Isaacs. Prostate cancer. N. Engl. J. Med 349:366–381 (2003).

    Article  PubMed  CAS  Google Scholar 

  3. N. Y. Chiu, and K. H. Chang. The illustrated medicinal plants of Taiwan. SMC, Taipei, 1986, p. 172.

    Google Scholar 

  4. Y. C. Wang, and T. L. Huang. Screening of anti-Helicobacter pylori herbs deriving from Taiwanese folk medicinal plants. FEMS Immunol. Med. Microbiol 43:295–300 (2005).

    Article  PubMed  CAS  Google Scholar 

  5. Y. C. Wang, and T. L. Huang. Anti-Helicobacter pylori activity of Plumbago zeylanica L. FEMS Immunol. Med. Microbiol 43:407–412 (2005).

    Article  PubMed  CAS  Google Scholar 

  6. Y. Dai, L. F. Hou, Y. P. Chan, L. Cheng, and P. P. But. Inhibition of immediate allergic reactions by ethanol extract from Plumbago zeylanica stems. Biol. Pharm. Bull 27:429–432 (2004).

    Article  PubMed  CAS  Google Scholar 

  7. M. J. Chan-Bacab, and L. M. Pena-Rodriguez. Plant natural products with leishmanicidal activity. Nat. Prod. Rep 18:674–688 (2001).

    Article  PubMed  CAS  Google Scholar 

  8. M. Krishnaswamy, and K. K. Purushothaman. Plumbagin: A study of its anticancer, antibacterial & antifungal properties. Indian J. Exp. Biol 18:876–877 (1980).

    PubMed  CAS  Google Scholar 

  9. I. Sharma, D. Gusain, and V. P. Dixit. Hypolipidaemic and antiatherosclerotic effects of plumbagin in rabbits. Indian J. Physiol. Pharmacol 35:10–14 (1991).

    PubMed  CAS  Google Scholar 

  10. R. Parimala, and P. Sachdanandam. Effect of Plumbagin on some glucose metabolizing enzymes studied in rats in experimental hepatoma. Mol. Cell. Biochem 125:59–63 (1993).

    Article  PubMed  CAS  Google Scholar 

  11. R. A. Naresh, N. Udupa, and P. U. Devi. Niosomal plumbagin with reduced toxicity and improved anticancer activity in BALB/C mice. J. Pharm. Pharmacol 48:1128–1132 (1996).

    PubMed  CAS  Google Scholar 

  12. S. Sugie, K. Okamoto, K. M. Rahman, T. Tanaka, K. Kawai, J. Yamahara, and H. Mori. Inhibitory effects of plumbagin and juglone on azoxymethane-induced intestinal carcinogenesis in rats. Cancer Lett 127:177–183 (1998).

    Article  PubMed  CAS  Google Scholar 

  13. P. Srinivas, G. Gopinath, A. Banerji, A. Dinakar, and G. Srinivas. Plumbagin induces reactive oxygen species, which mediate apoptosis in human cervical cancer cells. Mol. Carcinog 40:201–211 (2004).

    Article  PubMed  CAS  Google Scholar 

  14. P. L. Kuo, Y. L. Hsu, and C. Y. Cho. Plumbagin induces G2-M arrest and autophagy by inhibiting the AKT/mammalian target of rapamycin pathway in breast cancer cells. Molecular Cancer Therapeutics 5:3209–3221 (2006).

    Article  PubMed  Google Scholar 

  15. Y. L. Hsu, C. Y. Cho, P. L. Kuo, Y. T. Huang, and C. C. Lin. Plumbagin (5-hydroxy-2-methyl-1,4-naphthoquinone) induces apoptosis and cell cycle arrest in A549 cells through p53 accumulation via c-Jun NH2-terminal kinase-mediated phosphorylation at serine 15 in vitro and in vivo. J. Pharmacol. Exp. Ther 318:484–494 (2006).

    Article  PubMed  CAS  Google Scholar 

  16. P. U. Devi, B. S. Rao, and F. E. Solomon. Effect of plumbagin on the radiation induced cytogenetic and cell cycle changes in mouse Ehrlich ascites carcinoma in vivo. Indian J. Exp. Biol 36:891–895 (1998).

    PubMed  CAS  Google Scholar 

  17. A. Ganasoundari, S. M. Zare, and P. U. Devi. Modification of bone marrow radiosensensitivity by medicinal plant extracts. Br. J. Radiol 70:599–602 (1997).

    PubMed  CAS  Google Scholar 

  18. V. S. Prasad, P. U. Devi, B. S. Rao, and R. Kamath. Radiosensitizing effect of plumbagin on mouse melanoma cells grown in vitro. Indian J. Exp. Biol 34:857–858 (1996).

    PubMed  CAS  Google Scholar 

  19. S. K. Sandur, H. Ichikawa, G. Sethi, K. S. Ahn, and B. B. Aggarwal. Plumbagin (5-hydroxy-2-methyl-1,4-naphthoquinone) suppresses NF-kappaB activation and NF-kappaB-regulated gene products through modulation of p65 and IkappaBalpha kinase activation, leading to potentiation of apoptosis induced by cytokine and chemotherapeutic agents. J. Biol. Chem 281:17023–17033 (2006).

    Article  PubMed  CAS  Google Scholar 

  20. S. Miyamoto, and I. M. Verma. Rel/NF-kappa B/I kappa B story. Adv. Cancer Res 66:255–292 (1995).

    Article  PubMed  CAS  Google Scholar 

  21. Z. L. Chu, T. A. McKinsey, L. Liu, J. J. Gentry, M. H. Malim, and D. W. Ballard. 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 94:10057–10062 (1997).

    Article  PubMed  CAS  Google Scholar 

  22. M. You, P. T. Ku, R. Hrdlickova, and H. R. Bose. ch-IAP1, a member of the inhibitor-of-apoptosis protein family, is a mediator of the antiapoptotic activity of the v-Rel oncoprotein. Mol. Cell. Biol 17:7328–7341 (1997).

    PubMed  CAS  Google Scholar 

  23. W. X. Zong, L. C. Edelstein, C. Chen, J. Bash, and C. Gelinas. The prosurvival Bcl-2 homolog Bfl-1/A1 is a direct transcriptional target of NF-kappaB that blocks TNFalpha-induced apoptosis. Genes Dev 13:382–387 (1999).

    Article  PubMed  CAS  Google Scholar 

  24. L. Zhu, S. Fukuda, G. Cordis, D. K. Das, and N. Maulik. Anti-apoptotic protein survivin plays a significant role in tubular morphogenesis of human coronary arteriolar endothelial cells by hypoxic preconditioning. FEBS Lett 508:369–374 (2001).

    Article  PubMed  CAS  Google Scholar 

  25. R. C. Bargou, F. Emmerich, D. Krappmann, K. Bommert, M. Y. Mapara, W. Arnold, H. D. Royer, E. Grinstein, A. Greiner, C. Scheidereit, and B. Dörken. Constitutive nuclear factor-kappaB-RelA activation is required for proliferation and survival of Hodgkin's disease tumor cells. J. Clin. Invest 100:2961–2969 (1997).

    Article  PubMed  CAS  Google Scholar 

  26. R. L. Shattuck-Brandt, and A. Richmond. Enhanced degradation of I-kappaB alpha contributes to endogenous activation of NF-kappaB in Hs294T melanoma cells. Cancer Res 57:3032–3039 (1997).

    PubMed  CAS  Google Scholar 

  27. G. Dong, Z. Chen, T. Kato, and C. van Waes. The host environment promotes the constitutive activation of nuclear factor-kappaB and proinflammatory cytokine expression during metastatic tumor progression of murine squamous cell carcinoma. Cancer Res 59:3495–3504 (1999).

    PubMed  CAS  Google Scholar 

  28. S. T. Palayoor, M. Y. Youmell, S. K. Calderwood, C. N. Coleman, and B. D. Price. Constitutive activation of IkappaB kinase alpha and NF-kappaB in prostate cancer cells is inhibited by ibuprofen. Oncogene 18:7389–7394 (1999).

    Article  PubMed  CAS  Google Scholar 

  29. D. Xiao, S. Choi, D. E. Johnson, V. G., Vogel, C. S. Johnson, D. L. Trump, Y. J. Lee, and S. V. Singh. Diallyl trisulfide-induced apoptosis in human prostate cancer cells involves c-Jun N-terminal kinase and extracellular-signal regulated kinase-mediated phosphorylation of Bcl-2. Oncogene 23:5594–5606 (2004).

    Article  PubMed  CAS  Google Scholar 

  30. S. V. Singh, S. K. Srivastava, S. Choi, K. L. Lew, J. Antosiewicz, D. Xiao, Y. Zeng, S. C. Watkins, C. S. Johnson, D. L. Trump, Y. J. Lee, H. Xiao, and A. Herman-Antosiewicz. Sulforaphane-induced cell death in human prostate cancer cells is initiated by reactive oxygen species. J. Biol. Chem 280:19911–19924 (2005).

    Article  PubMed  CAS  Google Scholar 

  31. M. M. Webber, D. Bello, and S. Quader. Immortalized and tumorigenic adult human prostatic epithelial cell lines: characteristics and applications. Part 3. Oncogenes, suppressor genes, and applications. Prostate 30:136–142 (1997).

    Article  PubMed  CAS  Google Scholar 

  32. G. N. Thalmann, P. E. Anezinis, S. M. Chang, H. E. Zhau, E. E. Kim, V. L. Hopwood, S. Pathak, A. C. von Eschenbach, and L. W. Chung. Androgen-independent cancer progression and bone metastasis in the LNCaP model of human prostate cancer. Cancer Res 54:2577–2581 (1994).

    PubMed  CAS  Google Scholar 

  33. H. C. Wu, J. T. Hsieh, M. E. Gleave, N. M. Brown, S. Pathak, and L. W. Chung. Derivation of androgen-independent human LNCaP prostatic cancer cell sublines: role of bone stromal cells. Int. J. Cancer 57:406–412 (1994).

    Article  PubMed  CAS  Google Scholar 

  34. Y. J. Hsieh, L. C. Lin, and T. H. Tsai. Measurement and pharmacokinetic study of plumbagin in a conscious freely moving rat using liquid chromatography/tandem mass spectrometry. J. Chromatogr. B 844:1–5 (2006).

    Article  CAS  Google Scholar 

  35. D. Xiao, K. L. Lew, Y. Zeng, H. Xiao, S. W. Marynowski, R. Dhir, and S. V. Singh. Phenethyl isothiocyanate-induced apoptosis in PC-3 human prostate cancer cells is mediated by reactive oxygen species-dependent disruption of the mitochondrial membrane potential. Carcinogenesis 27:2223–2234 (2006).

    Article  PubMed  CAS  Google Scholar 

  36. D. Xiao, V. Vogel, and S. V. Singh. Benzyl isothiocyanate-induced apoptosis in human breast cancer cells is initiated by reactive oxygen species and regulated by Bax and Bak. Molecular Cancer Therapeutics 5:2931–2945 (2006).

    Article  PubMed  CAS  Google Scholar 

  37. Y. A. Kim, D. Xiao, H. Xiao, A. A. Powolny, K. L. Lew, M. L. Reilly, Y. Zeng, Z. Wang, and S. V. Singh. Mitochondria-mediated apoptosis by diallyl trisulfide in human prostate cancer cells is associated with generation of reactive oxygen species and regulated by Bax/Bak. Molecular Cancer Therapeutics 6:1599–1609 (2007).

    Article  PubMed  CAS  Google Scholar 

  38. S. V. Singh, S. Choi, Y. Zeng, E. Hahm, and D. Xiao. Guggulsterone-induced apoptosis in human prostate cancer cells is caused by reactive oxygen intermediate-dependent activation of c-Jun NH2-terminal kinase. Cancer Res 67:7439–7449 (2007).

    Article  PubMed  CAS  Google Scholar 

  39. L. J. Buccellato, M. Tso, O. I. Akinci, N. S. Chandel, and G. R. S. Budinger. Reactive oxygen species are required for hyperoxia-induced Bax activation and cell death in alveolar epithelial cells. J. Biol. Chem 279:6753–6760 (2004).

    Article  PubMed  CAS  Google Scholar 

  40. R. J. Davis. The mitogen-activated protein kinase signal transduction pathway. J. Biol. Chem 268:14553–14556 (1993).

    PubMed  CAS  Google Scholar 

  41. K. Lei, and R. J. Davis. JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis. Proc. Natl. Acad. Sci. U. S. A 100:2432–2437 (2003).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

This investigation was supported by the National Cancer Institute (NCI) grant CA115498.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shivendra V. Singh.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Powolny, A.A., Singh, S.V. Plumbagin-induced Apoptosis in Human Prostate Cancer Cells is Associated with Modulation of Cellular Redox Status and Generation of Reactive Oxygen Species. Pharm Res 25, 2171–2180 (2008). https://doi.org/10.1007/s11095-008-9533-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11095-008-9533-3

Key words

Navigation