Thromb Haemost 2009; 101(02): 373-380
DOI: 10.1160/TH08-01-0025
Cellular Proteolysis and Oncology
Schattauer GmbH

Treatment of PC3 prostate cancer cells with mitoxantrone, etoposide, doxorubicin and carboplatin induces distinct alterations in the expression of kallikreins 5 and 11

Hellinida Thomadaki
1   Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Athens, Panepistimiopolis, Athens, Greece
,
Konstantinos Mavridis
1   Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Athens, Panepistimiopolis, Athens, Greece
,
Maroulio Talieri
2   “G. Papanicolaou” Research Center of Oncology “Saint Savas” Hospital Athens, Greece
,
Andreas Scorilas
1   Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Athens, Panepistimiopolis, Athens, Greece
› Author Affiliations
Financial support: This work was supported by a Greek-Czech joint research and technology grant (EPAN.M.4.3.6.1) co-funded by the European Regional Development Fund (70%) and National Resources (30%) – Ministry of Development – General Secretariat for Research & Technology of Greece and by the Biopaths Co.
Further Information

Publication History

Received: 13 January 2008

Accepted after major revision: 01 January 2008

Publication Date:
23 November 2017 (online)

Summary

Several of the novel kallikrein-related peptidases (tissue kallikreins; KLKs) are emerging new serum and/or tissue biomarkers for prostate cancer (CaP) diagnosis, prognosis and monitoring. In the present research approach, our objective was to investigate the possible alterations in the mRNA expression levels of KLK5 and KLK11 genes in prostate cancer cells PC3 as a response to treatment with Mitoxantrone, Etoposide, Doxorubicin and Carboplatin. Viability was assessed with the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay after cell treatment with either mitoxantrone (2 μM), etoposide (20 μM), doxorubicin (1 μM), or carboplatin (15 μM), for 24, 48 and 72 hours. Additionally, trypan blue staining revealed that in PC3 cells all drugs displayed almost the same limited necrotic effects which appeared mainly at 72 hours of treatment. PC3 prostate cancer cells showed a concentration- and time-dependent increased cytotoxicity to the drugs under study which was mainly due to reduction of cell proliferation efficiency. Distinct modulations of KLK5 and KLK11 genes, at the mRNA level, were observed, supporting a drug-dependent cell response. Our experimental data demonstrate that the molecular profile mainly of KLK5 gene may serve as a new potential molecular biomarker predicting treatment response in CaP cells.

 
  • References

  • 1 Hsing AW, Chokkalingam AP. Prostate cancer epidemiology. Front Biosci 2006; 11: 1388-1413.
  • 2 Reynolds MA, Kastury K, Groskopf J. et al. Molecular markers for prostate cancer. Cancer Lett 2007; 249: 5-13.
  • 3 Rubin MA, De Marzo AM. Molecular genetics of human prostate cancer. Mod Pathol 2004; 17: 380-388.
  • 4 Nelson WG, Yegnasubramanian S, Agoston AT. et al. Abnormal DNA methylation, epigenetics, and prostate cancer. Front Biosci 2007; 12: 4254-4266.
  • 5 Shand RL, Gelmann EP. Molecular biology of prostate-cancer pathogenesis. Curr Opin Urol 2006; 16: 123-131.
  • 6 Dehm SM, Tindall DJ. Molecular regulation of androgen action in prostate cancer. J Cell Biochem 2006; 99: 333-344.
  • 7 Richter E, Srivastava S, Dobi A. Androgen receptor and prostate cancer. Prostate Cancer Prostatic Dis 2007; 10: 114-118.
  • 8 Collins AT, Maitland NJ. Prostate cancer stem cells. Eur J Cancer 2006; Jun 42 (09) 1213-8.
  • 9 Bradford TJ, Tomlins SA, Wang X. et al. Molecular markers of prostate cancer. Urol Oncol 2006; 24: 538-551.
  • 10 Magklara A, Scorilas A, Catalona WJ, Diamandis EP. The combination of human glandular kallikrein and free prostate-specific antigen (PSA) enhances discrimination between prostate cancer and benign prostatic hyperplasia in patients with moderately increased total PSA. Clin Chem, 1999; 45: 1960-1966.
  • 11 Yousef GM, Chang A, Scorilas A, Eleftherios P. Diamandis. Genomic organization of the human kallikrein locus on chromosome 19q13.3-q13.4. Biochem Bioph Res Commun, 2000; 276: 125-133.
  • 12 Borgono CA, Michael IP, Diamandis EP. Human tissue kallikreins: physiologic roles and applications in cancer. Mol Cancer Res 2004; 2: 257-280.
  • 13 Kurlender L, Borgono C, Michael IP. et al. A survey of alternative transcripts of human tissue kallikrein genes. Biochim Biophys Acta 2005; 1755: 1-14.
  • 14 Hooley E, McEwan PA, Emsley J. Molecular modeling of the prekallikrein structure provides insights into high-molecular-weight kininogen binding and zymogen activation. J Thromb Haemost 2007; 5: 2461-2466.
  • 15 Schmaier AH, McCrae KR. The plasma kallikreinkinin system: its evolution from contact activation. J Thromb Haemost 2007; 5: 2323-2329.
  • 16 Yousef GM, Diamandis EP. Expanded human tissue kallikrein family--a novel panel of cancer biomarkers. Tumour Biol. 2002; 23: 185-192.
  • 17 Obiezu CV, Diamandis EP. Human tissue kallikrein gene family: applications in cancer. Cancer Lett 2005; 224: 1-22.
  • 18 Paliouras M, Borgono C, Diamandis EP. Human tissue kallikreins: the cancer biomarker family. Cancer Lett 2007; 249: 61-79.
  • 19 Yousef GM, Diamandis EP. Human tissue kallikreins: a new enzymatic cascade pathway?. Biol Chem 2002; 383: 1045-1057.
  • 20 Yousef GM, Eleftherios PD. The new human tissue kallikrein gene family: structure, function, and association to disease. Endocrine Rev 2001; 22: 184-204.
  • 21 Yousef GM, Scorilas A, Diamandis EP. Genomic organization, mapping, tissue expression and hormonal regulation of trypsin-like serine protease (TLSP PRSS20), a new member of the human kallikrein gene family. Genomics 2000; 63: 88-96.
  • 22 Stavropoulou P, Gregorakis AK, Plebani M. et al. Expression analysis and prognostic significance of human kallikrein 11 in prostate cancer. Clin Chim Acta 2005; 357: 190-195.
  • 23 Nakamura T, Mitsui S, Okui A. et al. Alternative splicing isoforms of hippostasin [PRSS20/KLK11] in prostate cancer cell lines. Prostate 2001; 49: 72-78.
  • 24 Nakamura T, Stephan C, Scorilas A. et al. Quantitative analysis of hippostasin/KLK11 gene expression in cancerous and noncancerous prostatic tissues. Urology 2003; 61: 1042-1046.
  • 25 Borgono CA, Fracchioli S, Yousef GM. et al. Favorable prognostic value of tissue human kallikrein 11 [hK11] in patients with ovarian carcinoma. Int J Cancer 2003; 106: 605-610.
  • 26 Shigemasa K, Gu L, Tanimoto H. et al. Human kallikrein gene 11 [KLK11] mRNA overexpression is associated with poor prognosis in patients with epithelial ovarian cancer. Clin Cancer Res 2004; 10: 2766-2770.
  • 27 Petraki CD, Papanastasiou PA, Karavana VN. et al. Cellular distribution of human tissue kallikreins: immunohistochemical localization. Biol Chem 2006; 387: 653-663.
  • 28 Shaw JL, Diamandis EP. Distribution of 15 human kallikreins in tissues and biological fluids. Clin Chem 2007; 53: 1423-1432.
  • 29 Yousef GM, Diamandis EP. The new kallikrein-like gene, KLK-L2. Molecular characterization, mapping, tissue expression, and hormonal regulation. J Biol Chem 1999; 274: 37511-37516.
  • 30 Yousef GM, Scorilas A, Kyriakopoulou LG. et al. Human kallikrein gene 5 [KLK5] expression by quantitative PCR: an independent indicator of poor prognosis in breast cancer. Clin Chem 2002; 48: 1241-1250.
  • 31 Shinoda Y, Kozaki K, Imoto I. et al. Association of KLK5 overexpression with invasiveness of urinary bladder carcinoma cells. Cancer Sci 2007; 98: 1078-1086.
  • 32 Kim H, Scorilas A, Katsaros D. et al. Human kallikrein gene 5 [KLK5] expression is an indicator of poor prognosis in ovarian cancer. Br J Cancer 2001; 84: 643-650.
  • 33 Luo LY, Yousef G, Diamandis EP. Human tissue kallikreins and testicular cancer. APMIS 2003; 111 (01) 225-32 discussion 32-33.
  • 34 Planque C, de Monte M, Guyetant S. et al. KLK5 and KLK7, two members of the human tissue kallikrein family, are differentially expressed in lung cancer. Biochem Biophys Res Commun 2005; 329: 1260-1266.
  • 35 Yousef GM, Scorilas A, Chang A. et al. Down-regulation of the human kallikrein gene 5 [KLK5] in prostate cancer tissues. Prostate 2002; 51: 126-32.
  • 36 Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983; 65: 55-63.
  • 37 Freshney R. Culture of Animal Cells: A Manual of Basic Techniqu. Alan R. Liss, Inc.; New York: 1987. pp 117.
  • 38 Barry MA, Behnke CA, Eastman A. Activation of programmed cell death (apoptosis) by cisplatin, other anticancer drugs, toxins and hyperthermia. Biochem Pharmacol 1990; 40: 2353-2362.
  • 39 Fleming MT, Morris MJ, Heller G. et al. Post-therapy changes in PSA as an outcome measure in prostate cancer clinical trials. Nat Clin Pract Oncol 2006; 3: 658-67.
  • 40 Cheung R, Tucker SL, Kuban DA. First-year PSA kinetics and minima after prostate cancer radiotherapy are predictive of overall survival. Int J Radiat Oncol Biol Phys 2006; 66: 20-4.
  • 41 Keetch DW, Andriole GL, Ratliff TL. et al. Comparison of percent free prostate-specific antigen levels in men with benign prostatic hyperplasia treated with finasteride, terazosin, or watchful waiting. Urology 1997; 50: 901-5.
  • 42 Crook J, Gillan C, Yeung I. et al. PSA kinetics and PSA bounce following permanent seed prostate brachytherapy. Int J Radiat Oncol Biol Phys 2007; 69: 426-33.
  • 43 McLean M, McGowan TS, Catton CN. et al. The impact of prostatic manipulation upon serum PSA in patients with adenocarcinoma of the prostate. Clin Oncol (R Coll Radiol) 1993; 5: 293-6.
  • 44 Sridhara R, Eisenberger MA, Sinibaldi VJ. et al. Evaluation of prostate-specific antigen as a surrogate marker for response of hormone-refractory prostate cancer to suramin therapy. J Clin Oncol 1995; 13: 2944-53.
  • 45 Boland MP, Fitzgerald KA, O’Neill LA. Topoisomerase II is required for mitoxantrone to signal nuclear factor kappa B activation in HL60 cells. J Biol Chem 2000; 275: 25231-25238.
  • 46 Harker WG, Slade DL, Drake FH, Parr RL. Mitoxantrone resistance in HL-60 leukemia cells: reduced nuclear topoisomerase II catalytic activity and drug-induced DNA cleavage in association with reduced expression of the topoisomerase II beta isoform. Biochemistry 1991; 30: 9953-9961.
  • 47 van Maanen JM, Retel J, de Vries J, Pinedo HM. Mechanism of action of antitumor drug etoposide: a review. J Natl Cancer Inst 1988; 80: 1526-1533.
  • 48 Dwarakanath BS, Khaitan D, Mathur R. Inhibitors of topoisomerases as anticancer drugs: problems and prospects. Indian J Exp Biol 2004; 42: 649-659.
  • 49 Baldwin EL, Osheroff N. Etoposide, topoisomerase II and cancer. Curr Med Chem Anti-Cancer Agents 2005; 5: 363-372.
  • 50 Bose RN. Biomolecular targets for platinum antitumor drugs. Mini Rev Med Chem 2002; 2: 103-111.
  • 51 Floros KV, Thomadaki H, Lallas G, Katsaros N, Talieri M, Scorilas A. Cisplatin-induced apoptosis in HL-60 human promyelocytic leukaemia cells: differential expression of BCL2 and novel apoptosis-related gene BCL2L12. Ann NY Acad Sci 2003; 1010: 153-158.
  • 52 Floros KV, Thomadaki H, Katsaros N, Talieri M, Scorilas A. mRNA expression analysis of a variety of apoptosis-related genes, including the novel gene of the BCL2-family, BCL2L12, in HL-60 leukemia cells after treatment with carboplatin and doxorubicin. Biol Chem 2004; 385: 1099-1103.
  • 53 Floros KV, Talieri M, Scorilas A. Topotecan and methotrexate alter expression of the apoptosis-related genes BCL2, FAS and BCL2L12 in leukemic HL-60 cells. Biol Chem 2006; 387: 1629-1633.
  • 54 Huang R, Wallqvist A, Covell DG. Anticancer metal compounds in NCI’s tumor-screening database: putative mode of action. Biochem Pharmacol 2005; 69: 1009-1039.
  • 55 Hortobagyi GN. Chemotherapy of breast cancer: a historical perspective. Semin Oncol 1997; 17: S1-S4.