eISSN: 1897-4309
ISSN: 1428-2526
Contemporary Oncology/Współczesna Onkologia
Current issue Archive Manuscripts accepted About the journal Supplements Addendum Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
6/2015
vol. 19
 
Share:
Share:
Review paper

Role of microRNAs in the resistance of prostate cancer to docetaxel and paclitaxel

Ewa Kopczyńska

Contemp Oncol (Pozn) 2015; 19 (6): 423–427
Online publish date: 2016/01/13
Article file
- Role.pdf  [0.07 MB]
Get citation
 
PlumX metrics:
 

Introduction

The taxane class of cytotoxic chemotherapeutic agents has been shown to provide a survival benefit in advanced prostate cancer (PCa), inhibiting tumour development [1]. Taxanes (docetaxel, paclitaxel) suppress micro tubular depolymerization and attenuate the effects of bcl-2 and bcl-xL gene expression. Taxane-induced microtubule stabilization arrests cells in the G(2)M phase of the cell cycle and induces bcl-2 phosphorylation, thereby promoting a cascade of events that ultimately leads to apoptotic cell death [2]. Apart from the afore-mentioned, taxanes may affect androgen receptor (AR) signaling that they inhibit ligand-induced AR nuclear translocation and downstream transcriptional activation of AR target genes such as prostate-specific antigen [3].
Although taxanes inhibit tumour growth and improve survival in advanced PCa, development of resistance is inevitable and patients’ disease eventually progresses. Resistance can be formed through a variety of mechanisms; both intrinsic to prostate cancer biology (e.g. AR up regulation, increased extragonadal androgen synthesis) and general resistance mechanisms (limited tumor/tissue penetration, inherently resistant subpopulation of cells, multidrug resistance efflux pump) [4]. Increased transport of the drug out of tumour cells by up-regulation of ATP-binding cassette transporter molecules in the cell membrane, such as P-glycoprotein, is classified as a common resistance mechanism [1].
The mechanisms of docetaxel resistance include inhibition of apoptosis and activation of the extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) or phosphatidylinositol-3 kinase/Akt survival pathways as well. Additionally, docetaxel-resistant PCa cells exhibit an epithelial-mesenchymal transition (EMT) phenotype linking docetaxel resistance with the development of metastasis, whereas up-regulation of ZEB1 transcription factor, a direct regulator of EMT, can confer docetaxel resistance [1].
MicroRNAs (miRNAs) are small (~22 nt) no coding RNAs that regulate gene expression causing post-transcriptional inhibition or degradation of target mRNA. MiRNAs serve crucial components in the regulation of many cellular functions and biological processes [5]. Scientific studies have demonstrated that aberrant expressions of miRNAs, which can act as tumor suppressors or oncogenes, are closely associated with the development, invasion, metastasis and prognosis of various cancers including prostate cancer (PCa) [6]. The initial studies of miRNAs deregulation in prostate cancer were performed by miRNA microarray profiling and, since then, several researchers have analyzed prostate cancer specific miRNA profiles using genome-wide screenings and validation by quantitative PCR technology [7–15].
Nearly 50 miRNAs have been reported to be differentially expressed in human prostate cancer (about forty up- and remaining down-regulated) so far, though, knowledge concerning the effects of miRNAs on the sensitivity to anti-cancer drugs is still limited. Several groups of researches have studied whether the sensitivity to docetaxel and paclitaxel could be altered by miRNAs in prostate cancer cells. The development of chemoresistance has been attributed to alterations at the level of miRNAs. Table 1 presents the expression changes only for miRNAs involved in taxanes resistance. These results are not perfectly correlated as it could be caused by the application of different analytical methods.

miR-21 – oncomiR (role in docetaxel-resistance)

The miR-21 is one of the most commonly implicated miRNAs in cancer as its expression is highly up-regulated in a variety of solid tumors. In prostate cancer miR-21 is crucial in tumorigenesis and invasiveness (induction of tumor angiogenesis and initiation of epithelial-mesenchymal transition) by targeting PDCD4, PTEN, RECK, and BTG2. miR-21 can directly down-regulate the expression of PDCD4 (programmed cell death 4; neoplastic transformation inhibitor protein) – suppressor of tumorigenesis in PC3 cells [16]. Overexpression of miR-21 in DU145 cells increased the expression of HIF-1 and VEGF and induced tumor angiogenesis. MiR-21 induces tumor angiogenesis through targeting PTEN, leading to activate AKT and ERK1/2 signaling pathways and thereby, enhancing HIF-1 and VEGF expression; HIF-1 is a key downstream target of miR-21 in regulating tumor angiogenesis [17]. Apart from, in PCa cell line, DU-145 miR-21 directly inhibits RECK, a tumor suppressor gene involved in the control of matrix metalloproteinase 9 (MMP9) [18]. Coppola et al. [19] investigated whether down-regulation of the basal protein B-cell translocation gene 2 (BTG2) is implicated in prostate cancer transformation and progression. It was shown that BTG2 loss can shift normal prostate basal cells towards luminal markers expression, a phenotype also accompanied by the appearance of epithelial-mesenchymal transition (EMT) traits. Additionally, the research proved that the overexpression of miR-21 suppresses BTG2 levels and promotes the acquisition of luminal markers and EMT in prostate cells.
Using microarrays Shi et al. [20] found that a number of miRNAs were significantly altered in the docetaxel-resistant PC3 cells (PC3R). miR-21, one of the miRNAs identified by microarrays, was up-regulated in PC3R cells. Ectopic expression of miR-21 increased the resistance of PC3 to docetaxel in the wild type cells. In contrast, silencing of miR-21 with transient transfection of its inhibitors led to sensitize the cells to docetaxel. These findings suggest that miR-21 contributes to the resistance of PC3R cells to docetaxel. Researchers found that miR-21 can directly down-regulate the expression of PDCD4 – suppressor of tumorigenesis. Silencing of PDCD4 expression increased the cell viability and resistance to docetaxel in PC3 cells suggesting that PDCD4 is a functional target for miR-21 induced chemoresistance to docetaxel. MiR-21 functions as a significant regulator of prostate cancer cell resistance to docetaxel, which provides new evidence that miRNAs may be involved in the tumor resistance to chemotherapy.
In another study [21], miR-21 was up-regulated in docetaxel-resistant prostate cancer PC3R cells as well. Similarly to Shi’s et al. [20] results, ectopic expression of miR-21 increased the resistance to docetaxel in PC3 wild-type cells, while silencing of miR-21 in PC3R cells sensitized the cells to docetaxel. The authors [21] state that miR-21 is not per se a central player in the onset of PCa and that its single hitting does not represent a valuable therapeutic intervention in such a disease. Their findings contribute to support the theory that the oncogenic properties of miR-21 and generally speaking that of any miRNAs could be cell and tissue dependent and its potential role as a biomarker or therapeutic target should be put in the context of a given disease.
Additionally, Zhang et al. [22] discovered that serum miR-21 levels were higher in hormone refractory prostate cancer (HRPC) patients than those with androgen dependent prostate cancer (ADPC) and localized PCa. Serum miR-21 levels were higher in the HRPC patients resistant to docetaxel – based chemotherapy, when compared to those sensitive to chemotherapy. As a result, levels of serum miR-21 correlated to levels of serum PSA in patients with metastatic PCa. Their results have indicated that miR-21 may be a useful biomarker for patients with PCa during disease progression.

miR-34a – tumor suppressor (role in paclitaxel- and camptothecin-resistance)

The expression levels of miR-34a were markedly decreased in androgen-refractory PC3 and Du145 cells compared to androgen-sensitive LNPCa and normal prostate epithelial cells. Furthermore, miR-34a expression depends on the p53 activity in prostate cancer cell lines and appears to be completely absent in p53-null PC3 cells. The expression level of silent mating type information regulation 2 homolog 1 (SIRT1) was up regulated in p53-defective PC3 and DU145 cell. SIRT1 deacetylates pro-apoptotic proteins such as p53 and promotes cell survival under genotoxic and oxidative stress. The anti-apoptotic activity of SIRT1 is implicated in tumorigenesis. In addition, SIRT1 is suggested to be involved in resistance to anticancer drug. In p53-null PC3 cells, introduction of p53 increased miR-34a expression. Ectopic expression which in turn reduced SIRT1 expression. It is therefore presumed that miR-34a can inhibit cell growth and enhance chemosensitivity to camptothecin [23].
Kojima et al. [24] have explained miR-34 action. The miR directly and indirectly via regulating HuR expression acts on the 3’-UTR of SIRT1 and Bcl2 mRNAs and suppresses their expression. Decreased expression of miR-34a leads to the up-regulation of SIRT1 and Bcl2, resulting in resistance to apoptosis caused by paclitaxel.
MicroRNA profiling of DU14-TXR and PC3-TXR cells and prostate cancer tissue from the patients done by Singh et al. [25] showed decreased expression of miR-34a. Experimental replenishment of miR 34a in cultured cells prevents metastatsis and invasion. They concluded that chemoresistance to paclitaxel in DU145-TXR and PC3-TXR cells is possibly regulated by miRNAs, which are differentially expressed when the paclitaxel sensitive cell line is transformed to a resistant phenotype.

miR 143 – tumor suppressor (role in docetaxel-resistance)

Xu et al. [26] observed an inverse correlation of expression between miR-143 and KRAS protein (V Ki ras2 Kirsten rat sarcoma viral oncogene homolog) in prostate cancer cell lines. Oncogene KRAS is the key molecule of EGFR/RAS/MAPK signaling way which regulates a variety of biological activities, including cell proliferation, migration and chemosensitivity. In addition to KRAS, ERK5 (extracellular signal-regulated kinase 5) becomes also a target of miR-143. Ahmad’s et al. [27] findings show a significant correlation between low miR 143 and elevated ERK5 levels in primary human prostate cancers. MiR-143 contributes to suppressing tumor cell growth. Over-expression of miR-143 strikingly inhibited migration of prostate cancer cells in vitro. MiR-143-treated prostate cancer cells showed higher chemosensitivity to docetaxel, proving that miR-143 enhanced their response to docetaxel [26].

miR-148a – tumor suppressor (role in paclitaxel-resistance)

Fujita et al. [28] demonstrated that miR-148a is down-regulated in prostate cancer PC3 cells and DU-145-hormone refractory prostate cancer cells. A direct target of miR-148a in PC3 cells is mitogen- and stress-activated kinase 1 (MSK1). Ectopic expression of miR-148a decreased expression of MSK1 and inhibited growth, migration and invasion of PC3 cells. MiR-148a functions as a tumor suppressor. In paclitaxel-resistant cell line from PC3 cells (PC3PR), miR-148a attenuated the resistance to paclitaxel. The authors concluded that miR-148a is a promising therapeutic target for hormone-refractory prostate cancer especially for drug resistance prostate cancer.

miR-200 family – tumor suppressors (role in docetaxel- and paclitaxel-resistance)

Members of the miR-200 family could inhibit epithelial-mesenchymal transition (EMT) and suppress cancer invasion by the direct repression of the translation factors zinc-finger E-boxbinding homeobox 1 and 2 (ZEB1 and ZEB2). MiR-200b is identified as a critical regulator of tumor invasion, metastasis, and chemosensitivity [29].
miR-200b was down regulated in clinic prostatic tumors and in PCa cell lines. Enforced miR-200b expression suppressed PCa cell proliferation and migration and enhanced chemosensitivity to docetaxel by targeting B-cell-specific Moloney murine leukemia virus insertion site 1 (Bmi-1). Bmi-1 was detected at higher levels in PCa and knockdown of Bmi-1 showed similar effects as miR-200b overexpression in PCa cells [30].
Screening for key regulators of an epithelial phenotype revealed a significantly reduced expression of miR-200c in docetaxel-resistant cells. A prolonged treatment with miRNAs resulted in elevated E-cadherin protein levels and an increase in the percentage of apoptotic cells [31].
MiR200c maintains ‘epithelialness’ of cancer cells by preventing endothelial mesenchymal transition and the assumption of an aggressive chemoresistant mesenchymal phenotype [25].

miR-205 and miR-31 – tumor suppressors (role in docetaxel-resistance)

Bhatnagar et al. [32] demonstrated that miR-205 and miR-31 are down-regulated in advanced prostate cancer cells. They identified Bcl-w as the potential target for miR-205, and E2F6 as the potential target for miR-31. Bcl-w is an antiapoptotic member of the Bcl-2 family proteins, whereas E2F6 inhibits UV- and hypoxia-induced apoptosis. The antiapoptotic properties of Bcl-w and E2F6 make them interesting targets for miR-205 and miR-31. Overexpression of miR-205 and miR-31 down-regulated Bcl-w and E2F6, respectively. Conversely, transfection of WPE1-NA22 cells with anti-miR miRNA inhibitors specific to miR-205 and miR-31 increased the protein levels of Bcl-w and E2F6, respectively. The anti-miR-205 and anti-miR-31 inhibitors also blocked docetaxel-induced apoptosis in WPE1-NA22 cells. Having been treated with docetaxel or cisplatin, cells stably expressing Bcl-w were significantly resistant to drug-induced apoptosis, comparing with empty vector-transfected cells. Similarly, cells stably expressing E2F6 were also resistant to docetaxel and cisplatin.

Let-7c

The let-7 family represents complex group of miRNAs that act as tumor suppressors. Let-7 a, b, c, d, f, γ were downregulated in prostate cancer and let-7 b, d, i – upregulated [33].
Nadminty’s et al. [34, 35] data showed that the levels of let-7c were lower in the castration-resistant cell lines C4-2B, LNCaP-s17 and LN-IL6. Let-7c suppresses prostate tumor growth by several pathways including regulation of IL-6, Myc, Lin28 and the AR.
Downregulation of let-7 leads to the activation of protein synthesis of IMP-1 (insulin-like growth factor mRNA binding protein 1) which increases levels of MDR1 (multi-drug resistance-1) protein in cancer cells. MDR1 acting as a transporter of microtubule targeting drugs (anthracyclines and taxanes) from intra- to extra-cellular spaces, is involved in chemoresistance [36].

The author declares no conflict of interest.

References

1. Fitzpatrick JM, de Wit R. Taxane mechanisms of action: potential implications for treatment sequencing in metastatic castration-resistant prostate cancer. Eur Urol 2014; 65: 1198-204.
2. Pienta KJ. Preclinical mechanisms of action of docetaxel and docetaxel combinations in prostate cancer. Semin Oncol 2001; 4 Suppl 15: 3-7.
3. Darshan MS, Loftus MS Thadani-Mulero M, et al. Taxane-induced blockade to nuclear accumulation of the androgen receptor predicts clinical responses in metastatic prostate cancer. Cancer Res 2011; 71: 6019-29.
4. Schweizer MT, Antonarakis ES. Chemotherapy and its evolving role in the management of advanced prostate cancer. Asian J Androl 2014; 16: 334-40.
5. Bartel DP. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004; 116: 281-97.
6. Iorio MV, Croce1 CM. microRNA involvement in human cancer. Carcinogenesis 2012; 33: 1126-33.
7. Walter BA, Valera VA, Pinto PA, Merino MJ. Comprehensive microRNA profiling of prostate cancer. J Cancer 2013; 4: 350-7.
8. Wach S, Nolte E, Szczyrba J, et al. MicroRNA profiles of prostate carcinoma detected by multiplatform microRNA screening. Int J Cancer 2012; 130: 611-21.
9. Szczyrba J, Löprich E, Wach S, et al. The MicroRNA profile of prostate carcinoma obtained by deep sequencing. Mol Cancer Res 2010; 8: 529-38.
10. Tong AW, Fulgham P, Jay C, et al. MicroRNA profile analysis of human prostate cancers. Cancer Gene Therapy 2009; 16: 206-16.
11. Ambs S, Prueitt RL, Yi M, et al. Genomic profiling of microRNA and messenger RNA reveals deregulated microRNA expression in prostate cancer. Cancer Res 2008; 68: 6162-70.
12. Porkka KP, Pfeiffer MJ, Waltering KK, Vessella RL, Tammela TLJ, Visakorpi T. MicroRNA expression profiling in prostate cancer. Cancer Res 2007; 67: 13.
13. Volinia S, Calin GA, Liu CG, et al microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 2006; 103: 2257-61.
14. Ozen M, Creighton CJ, Ozdemir M, Ittmann M. Widespread deregulation of microRNA expression in human prostate cancer. Oncogene 2008; 27: 1788-93.
15. Lu J, Getz G, Miska EA, et al. MicroRNA expression profiles classify human cancers. Nature 2005; 435.
16. Lu Z, Liu M, Stribinskis V, Klinge CM, Ramos KS, Colburn NH, Li Y. MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene 2008; 27: 4373-9.
17. Liu LZ, Li C, Chen Q. MiR-21 induced angiogenesis through AKT and ERK activation and HIF-1a expression. PLoS One 2011; 6: 1-9.
18. Reis ST, Pontes-Junior J, Antunes AA, et al. miR-21 may acts as an oncomir by targeting RECK, a matrix metalloproteinase regulator, in prostate cancer. BMC Urology 2012; 12: 1-7.
19. Coppola V, Musumeci M, Patrizii M, et al. BTG2 loss and miR-21 up-regulation contribute to prostate cell transformation by inducing luminal markers expression and epithelial-mesenchymal transition. Oncogene 2013; 32: 1843-53.
20. Shi G, Ye D, Yao X, et al. Involvement of microRNA-21 in mediating chemoresistance to docetaxel in androgen-independent prostate cancer PC3 cells. Acta Pharmacol. Sinica 2010; 31: 867-73.
21. Folini M, Gandellini P, Longoni N, et al. miR-21: an oncomir on strike in prostate cancer. Molecular Cancer 2010; 9: 12.
22. Zhang HL, Yang LF, Zhu Y, et al. Serum miRNA-21: elevated levels in patients with metastatic hormone-refractory prostate cancer and potential predictive factor for the efficacy of docetaxel-based chemotherapy. Prostate 2011; 71: 326 331.
23. Fujita Y, Kojima K, Hamada N, Ohhashi R, Akao Y, Nozawa Y, Deguchi T, Ito M. Effects of miR-34a on cell growth and chemoresistance in prostate cancer PC3 cells. Biochem Biophys Res Commun 2008; 377: 114-9.
24. Kojima K, Fujita Y, Nozawa Y, Deguchi T, Ito M. MiR 34a attenuates paclitaxel-resistance of hormone-refractory prostate cancer PC3Cells through direct and indirect mechanisms. Prostate 2010; 70: 1501-12.
25. Singh S, Chitkara D, Mehrazin R, Behrman SW, Wake RW, Mahat RI. Chemoresistance in prostate cancer cells is regulated by miRNAs and hedgehog pathway. PLoS One 2012; 7: 6.
26. Xu B, Niu X, Zhang X, et al. miR-143 decreases prostate cancer cells proliferation and migration and enhances their sensitivity to docetaxel through suppression of KRAS. Mol Cell Biochem 2011; 350: 207-13.
27. Ahmad I, Singh LB, Yang ZH, et al. MiR143 expression inversely correlates with nuclear ERK5 immunoreactivity in clinical prostate cancer. Br J Cancer 2013; 108: 149-54.
28. Fujita Y, Kojima K, Ohhashi R, et al. MiR-148a attenuates paclitaxel resistance of hormone-refractory, drug-resistant prostate cancer PC3 cells by regulating MSK1 expression. J Biol Chem 2010; 285: 19076-84.
29. Feng B, Wang R, Chen LB. Review of MiR-200b and cancer chemosensitivity. Biomed Pharmacother 2012; 66: 397-402.
30. Yu J, Lu Y, Cui D, Li E, Zhu Y, Zhao Y, Zhao F, Xia S. miR-200b suppresses cell proliferation, migration and enhances chemosensitivity in prostate cancer by regulating Bmi-1. Oncol Rep 2014; 31: 910-8.
31. Puhr M, Hoefer J, Schäfer G, Erb H, Oh SJ, Klocker H, Heidegger I, Neuwirt H, Culig Z. Epithelial-to-mesenchymal transition leads to docetaxel resistance in prostate cancer and is mediated by reduced expression of miR-200c and miR-205. Am J Pathol 2012; 181: 6.
32. Bhatnagar N, Li X, Padi SKR, Zhang Q, Tang MS, Guo B. Downregulation of miR-205 and miR-31 confers resistance to chemotherapy-induced apoptosis in prostate cancer cells. Cell Death Dis 2010; 1: 1-8.
33. Chiu SC, Chung HY, Cho DY, et al. Therapeutic potential of microRNA let-7: tumor suppression or impeding normal stemness. Cell Transplant 2014; 23: 459-69.
34. Nadiminty N, Tummala R, Lou W, et al. MicroRNA let-7c suppresses androgen receptor expression and activity via regulation of Myc expression in prostate cancer cells. J Biol Chem 2012; 287: 1527-37.
35. Nadiminty N, Tummala R, Lou W, et al. MicroRNA let-7c is downregulated in prostate cancer and suppresses prostate cancer growth. PLoS ONE 2012; 7: e32832.
36. Pekarik V, Gumulec J, Masarik M, Kizek R, Adam V. Prostate cancer, miRNAs, metallothioneins and resistance to cytostatic drugs. Curr Med Chem 2013; 20: 534-44.

Address for correspondence

Ewa Kopczyńska
Department of Pathobiochemistry and Clinical Chemistry
Collegium Medicum in Bydgoszcz
M. Curie-Skłodowskiej 9
85-094 Bydgoszcz, Poland
e-mail: kopczynska@cm.umk.pl

Submitted: 4.07.2014
Accepted: 20.10.2014
Copyright: © 2016 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.