Open Access Journal Article

Effect of interleukin-8 on docetaxel resistance in prostate cancer cells: insights into the role of multidrug resistance 1 protein modulation

by Suleyman ILHAN a,* orcid
a
Manisa Celal Bayar University, Faculty of Science and Letters, Biology Department Manisa,Türkiye
*
Author to whom correspondence should be addressed.
CI  2023, 18; 2(1), 18; https://doi.org/10.58567/ci02010004
Received: 20 May 2023 / Accepted: 7 June 2023 / Published Online: 14 June 2023

Abstract

Although docetaxel treatment yields a high survival rate for prostate cancer (PCa), resistance eventually develops in many patients. Understanding the underlying mechanisms of docetaxel resistance is essential for improving treatment strategies. Cytokines, which play a role in cell signaling and immune responses, have been implicated in drug resistance mechanisms. The study revealed that interleukin-8 (IL-8) was consistently overexpressed in both docetaxel-resistant PCa cell lines. Thus, the expression levels of cytokines released from docetaxel-sensitive (PC-3- and DU-145) and resistant (PC-3/R-DU-145/R) PCa cells were compared. IL-8 was found to be commonly expressed in resistant cell lines. This finding led to the hypothesis that IL-8 could play a key role in mediating PCa cell resistance to docetaxel. IL-8 siRNA treatment increased docetaxel sensitivity in both resistant cells. To demonstrate the mechanism of IL-8-related resistance, MDR1 expression was evaluated. After IL-8 siRNA treatment MDR1 expression was reduced in both resistant cells suggesting that IL-8 regulates the docetaxel resistance of PCa cells via modulation of multidrug resistance 1 (MDR1). By expanding the knowledge of the cytokines and their effect mechanisms, novel approaches can be developed for the treatment of docetaxel-resistant prostate cancer. Further investigations into the role of IL-8 in docetaxel resistance could offer valuable insights into the development of effective treatment strategies for PCa patients.


Copyright: © 2023 by ILHAN. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY) (Creative Commons Attribution 4.0 International License). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
Show Figures

Share and Cite

ACS Style
ILHAN, S. Effect of interleukin-8 on docetaxel resistance in prostate cancer cells: insights into the role of multidrug resistance 1 protein modulation. Cancer Insight, 2023, 2, 18. https://doi.org/10.58567/ci02010004
AMA Style
ILHAN S. Effect of interleukin-8 on docetaxel resistance in prostate cancer cells: insights into the role of multidrug resistance 1 protein modulation. Cancer Insight; 2023, 2(1):18. https://doi.org/10.58567/ci02010004
Chicago/Turabian Style
ILHAN, Suleyman 2023. "Effect of interleukin-8 on docetaxel resistance in prostate cancer cells: insights into the role of multidrug resistance 1 protein modulation" Cancer Insight 2, no.1:18. https://doi.org/10.58567/ci02010004
APA style
ILHAN, S. (2023). Effect of interleukin-8 on docetaxel resistance in prostate cancer cells: insights into the role of multidrug resistance 1 protein modulation. Cancer Insight, 2(1), 18. https://doi.org/10.58567/ci02010004

Article Metrics

Article Access Statistics

References

  1. Siegel, R.L., Miller, K.D., Jemal, A., (2020). Cancer statistics, 2020. CA: A Cancer Journal for Clinician https://doi.org/10.3322/caac.21590
  2. Trewartha, D., Carter, K., (2013). Advances in prostate cancer treatment. Nature Publishing Group 12, 823 –824. https://doi.org/10.1038/nrd4068
  3. Litwin, M.S., Tan, H.J., (2017). The diagnosis and treatment of prostate cancer: A review. JAMA -Journal of the
  4. Clarke, S.J., Rivory, L.P., (1999). Clinical pharmacokinetics of docetaxel. Clinical Pharmacokinetics 36, 99 –114. https://doi.org/10.2165/00003088-199936020-00002
  5. Gligorov, J.,Lotz, J.P., (2004). Preclinical Pharmacology of the Taxanes: Implications of the Differences. Oncologist 9, 3–8.https://doi.org/10.1634/theoncologist.9-suppl_2-3
  6. Hanauske, A.R., Degen, D., Hilsenbeck, S.G., Bissery, M.C., Von Hoff, D.D., (1992). Effects of taxotere and taxol on in vitro colony formation of freshly explanted human tumor cells. Anticancer Drugs 3, 121 –124. https://doi.org/10.1097/00001813-199204000-00008
  7. Aragon-Ching, J.B., Dahut, W.L., (2007). Chemotherapy in Androgen-Independent Prostate Cancer (AIPC): What ’s next after taxane progression? Cancer Therapy 5A, 151 –160
  8. Saad, F., Hotte, S.J., (2010). Guidelines for the management of castrate-resistant prostate cancer. Journal of the Canadian Urological Association 4,380 –384.
  9. Stone, K.R., Mickey, D.D., Wunderli, H., Mickey, G.H., Paulson, D.F., (1978). Isolation of ahuman prostate carcinoma cell line (DU 145). International Journal of Cancer 21, 274 –281. https://doi.org/10.1002/ijc.2910210305
  10. Kaighn, M.E., Narayan, K.S., Ohnuki, Y., Lechner, J.F., Jones, L.W., (1979). Establishment and characterization of a human prostatic carcinoma cell line (PC-3). Investigative Urology 17, 16 –23
  11. Liu, C., Zhu, Y., Lou, W., Nadiminty, N., Chen, X., Zhou, Q., Shi, X.B., deVere White, R.W., Gao, A.C., (2013). Functional p53 determines docetaxel sensitivity in prostate cancer cells. Prostate 73, 418 –427. https://doi.org/10.1002/pros.22583
  12. Skj øth, I.H.E., Issinger, O.G., (2006). Profiling of signaling molecules in four different human prostate carcinoma cell lines before and after induction of apoptosis. International Journal of Oncology 28, 217 –229. https://doi.org/10.3892/ijo.28.1.217
  13. Nakamoto, T., Chang, C., Li, A., Chodak2, G.W., (1992). Basic Fibroblast Growth Factor in Human Prostate Cancer Cells1. Cancer Research 52, 571 –577
  14. Frønsdal, K., Saatcioglu, F., (2005). Histone deacetylase inhibitors differentially mediate apoptosis in prostate cancer cells. Prostate 62, 299 –306. https://doi.org/10.1002/pros.20140
  15. Alimirah, F., Chen, J., Basrawala, Z., Xin, H., Choubey, D., (2006). DU-145 and PC-3 human prostate cancer cell lines express androgen receptor: Implications for the androgen receptor functions and regulation. FEBS Letters 580, 2294 –2300. https://doi.org/10.1016/j.febslet.2006.03.041
  16. Erten, C., Karaca, B., Kucukzeybek, Y., Gorumlu, G., Cengiz, E., Gul, M.K., Atmaca, H., Uzunoglu, S., Karabulut, B., Sanli, U.A., Uslu, R., (2009). Regulation of growth factors in hormone- and drug-resistant prostate cancer cells by synergistic combination of docetaxel and octreotide. BJU International 104, 107 –114. https://doi.org/10.1111/j.1464-410X.2009.08340.x
  17. Jayakumar, S., Kunwar, A., Sandur, S.K., Pandey, B.N., Chaubey, R.C., (2014). Differential response of DU145 and PC3 prostate cancer cells to ionizing radiation: Role of reactive oxygen species, GSH and Nrf2 in radiosensitivity. Biochimica et Biophysica Acta -General Subjects 1840, 485 –494. https://doi.org/10.1016/j.bbagen.2013.10.006
  18. Alvin Y. Liu, (2000). Differential Expression of Cell Surface Molecules in Prostate Cancer Cells |Cancer Research. Cancer Research 60, 3429-3434
  19. Hwang, C., (2012). Overcoming docetaxel resistance in prostate cancer: Aperspective review. Therapeutic Advances in Medical Oncology 4,329 –340. https://doi.org/10.1177/1758834012449685
  20. Cengiz, E., Karaca, B., Kucukzeybek, Y., Gorumlu, G., Gul, M.K., Erten, C., Atmaca, H., Uzunoglu, S., Karabulut, B., Sanli, U.A., Uslu, R., (2010). Overcoming drug resistance in hormone-and drug-refractory prostate cancer cell line, PC-3 by docetaxel and gossypol combination. Molecular Biology Reports 37, 1269 –1277.
  21. Ganju, A., Yallapu, M.M., Khan, S., Behrman, S.W., Chauhan, S.C., Jaggi, M., (2014). Nanoways to overcome docetaxel resistance in prostate cancer. Drug Resistance Updates , Churchill Livingstone, pp. 13 –23. https://doi.org/10.1016/j.drup.2014.04.001
  22. Bhangal, G., Halford, S., Wang, J., Roylance, R., Shah, R., Waxman, J., (2000). Expression of the multidrug resistance gene in human prostate cancer. Urologic Oncology 5,118 –121. https://doi.org/10.1016/S1078-1439(99)00055-1
  23. Soto-Ortiz, L., (2016). Acancer treatment based on synergy between anti-angiogenic and immune cell therapies. Journal of Theoretical Biology 394, 197 –211. https://doi.org/10.1016/j.jtbi.2016.01.026
  24. Dudek, A.Z., Mahaseth, H., (2005). Circulating Angiogenic Cytokines in Patients with Advanced Non-Small Cell Lung Cancer: Correlation with Treatment Response and Survival. Cancer Investigation 23, 193 –200. https://doi.org/10.1081/CNV-200055949
  25. Neufeld, G., Kessler, O., (2006). Pro-angiogenic cytokines and their role in tumor angiogenesis. Cancer and Metastasis Reviews. https://doi.org/10.1007/s10555-006-9011-5
  26. Reiland, J., Furcht, L.T., McCarthy, J.B., (1999). CXC-chemokines stimulate invasion and chemotaxis in prostate carcinoma cells through the CXCR2 receptor. Prostate 41, 78 –88. https://doi.org/10.1002/(SICI)1097-0045(19991001)41:2<78::AID-PROS2>3.0.CO;2-P
  27. Lu, Y., Cai, Z., Galson, D.L., Xiao, G., Liu, Y., George, D.E., Melhem, M.F., Yao, Z., Zhang, J.,(2006). Monocyte chemotactic protein-1 (MCP-1) acts as aparacrine and autocrine factor for prostate cancer growth and invasion. Prostate 66, 1311 –1318. https://doi.org/10.1002/pros.20464
  28. Ries, C., (2014). Cytokine functions of TIMP-1. Cellular and Molecular Life Sciences .Springer, pp. 659 –672. https://doi.org/10.1007/s00018-013-1457-3
  29. Guccini, I.,Revandkar, A., D’Ambrosio, M., Colucci, M., Pasquini, E., Mosole, S., Troiani, M., Brina, D., Sheibani-Tezerji, R., Elia, A.R., Rinaldi, A., Pernigoni, N., Rüschoff, J.H., Dettwiler, S., De Marzo, A.M., Antonarakis, E.S., Borrelli, C., Moor, A.E., Garcia-Escudero, R., Alajati, A., Attanasio, G., Losa, M., Moch, H., Wild, P., Egger, G., Alimonti, A., (2021). Senescence Reprogramming by TIMP1 Deficiency Promotes Prostate Cancer Metastasis. Cancer Cell 39, 68-82.e9.
  30. Powell, W.C., Knox, J.D., Navre, M., Grogan, T.M., Kittelson, J., Nagle, R.B., Bowden, G.T., (1993). Expression of the Metalloproteinase Matrilysin in DU-145 Cells Increases Their Invasive Potential in Severe Combined Immunodeficient Mice. Cancer Research 53 (2)
  31. Hébert, C.A., Baker, J.B., (1993). Interleukin-8: A review. Cancer Investigation Informa Healthcare, pp. 743 –75 https://doi.org/10.3109/07357909309046949
  32. Matsushima, K., Baldwin, E.T., Mukaida, N., (1992). Interleukin-8 and MCAF: Novel Leukocyte Recruitment and Activating Cytokines (Part 2of 2), in: Interleukins: Molecular Biology and Immunology .KARGER, Basel, pp. 251 –265. https://doi.org/10.1159/000319091
  33. Campbell, L.M., Maxwell, P.J., Waugh, D.J.J., (2013). Rationale and means to target pro-inflammatory interleukin-8 (CXCL8) signaling in cancer. Pharmaceuticals 6,929-959. https://doi.org/10.3390/ph6080929
  34. Kadkol, H., Jain, V., Patil, A.B., (2019). Multi drug resistance in cancer therapy-an overview. Journal of Critical Reviews 6,1–6.
  35. Shao, N., Chen, L.H., Ye, R.Y., Lin, Y., Wang, S.M., (2013). The depletion of Interleukin-8 causes cell cycle arrest and increases the efficacy of docetaxel in breast cancer cells. Biochemical and Biophysical Research Communications 431, 535 –541. https://doi.org/10.1016/j.bbrc.2013.01.022
  36. Codony-Servat, J., Mar ín-Aguilera, M., Visa, L., Garc ía-Alb éniz, X., Pineda, E., Fern ández, P.L., Filella, X., Gasc ón, P., Mellado, B., (2013). Nuclear factor-kappa Band interleukin-6 related docetaxel resistance in castration-resistant prostate cancer. Prostate 73, 512-21. https://doi.org/10.1002/pros.22591
  37. Du, J., He, Y., Li, P., Wu, W., Chen, Y., Ruan, H., (2018). IL-8 regulates the doxorubicin resistance of colorectal cancer cells via modulation of multidrug resistance 1(MDR1). Cancer Chemotherapy and Pharmacology 8, 1111 –1119. https://doi.org/10.1007/s00280-018-3584-x