Open Access

Effects of cisplatin on the LSD1-mediated invasion and metastasis of prostate cancer cells

Retraction in: /10.3892/mmr.2023.13027

  • Authors:
    • Zhi‑Yuan Chen
    • Hui Chen
    • Tao Qiu
    • Xiao‑Dong Weng
    • Jia Guo
    • Lei Wang
    • Xiu‑Heng Liu
  • View Affiliations

  • Published online on: July 28, 2016     https://doi.org/10.3892/mmr.2016.5571
  • Pages: 2511-2517
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer poses a major public health problem in men. Metastatic prostate cancer is incurable, and ultimately threatens the life of patients. Lysine‑specific demethylase 1 (LSD1) is an androgen receptor‑interacting protein that exerts a key role in regulating gene expression and is involved in numerous biological processes associated with prostate cancer. Cisplatin, also known as cis‑diamminedichloroplatinum or DDP, is a standard chemotherapeutic agent used to treat prostate cancer; however, it has the disadvantage of various serious side effects. The present study aimed to investigate the effects of LSD1 knockdown, and the interplay between LSD1 and DDP, on prostate cancer cell proliferation, apoptosis and invasion, and, therefore, the potential of LSD1 as a target for prostate cancer therapy. Flow cytometric analysis, Cell Counting kit 8 assay, Transwell assay and western blotting results revealed that LSD1 knockdown, in combination with DDP treatment, exerted antiproliferative, proapoptotic and anti‑invasive effects on PC3 prostate cancer cells. In addition, knockdown of LSD1 acted synergistically with DDP, thereby enhancing the induction of apoptosis, and the inhibition of proliferation and invasion in prostate cancer cells. These results indicated that LSD1 may serve as a potential therapeutic target, and may enhance the sensitivity of PC3 cells to DDP.

Introduction

Prostate cancer, which is characterized by the development of prostate epithelial malignant tumors, occurs solely in men and is associated with the highest rates of morbidity and mortality (1). Prostate cancer poses a major public health problem worldwide (1). The incidence of prostate cancer is particularly prevalent in older men (2). The therapeutic strategies currently used to treat prostate cancer include watchful waiting, surgery, radiotherapy, chemotherapy, hormone therapy and biotherapy (3). The androgen receptor (AR) has an important role in prostate cancer development and progression (3). Androgen-deprivation therapy (ADT) is an important means of treatment for patients with prostate cancer; however, one disadvantage is that the prostate cancer may develop resistance to ADT over time (4). At present, there is a dearth of effective treatment methods which are beneficial to those patients who have developed androgen resistance in prostate cancer. Therefore, prostate cancer therapy remains unsatisfactory, and there is an urgent requirement to identify novel therapeutic strategies to overcome resistance to androgens in patients with prostate cancer.

Lysine specific demethylase 1 (LSD1) is a histone demethylase, which exerts important roles in tumorigenesis (58). LSD1 has been reported to be highly expressed in various cancer cell types, particularly in prostate cancer (9). Previous studies have demonstrated that LSD1, as an AR-interacting protein, may promote AR-dependent gene expression, which subsequently leads to the constitutive maintenance of cancer cells via growth signals and an enhanced risk of tumor relapse (9,10). In addition, it has been suggested that histone modification patterns may be used to predict the risk of prostate cancer recurrence (11). Although LSD1 regulates the expression of a wide range of genes and is involved in the processes of prostate cancer progression and deterioration (9), the underlying molecular mechanisms remain to be fully elucidated. Therefore, the inhibition of LSD1 activity may provide a useful target for the treatment of prostate cancer.

Cisplatin, also known as cis-diamminedichloroplatinum or DDP, is a platinum-based drug commonly used in the clinic as a chemotherapeutic agent. It has numerous characteristic properties, including broad-spectrum anticancer activity and curative effects, which render it useful for the clinical treatment of various tumors (12). However, its use is associated with several side effects, which serve to limit the doses that may be administered, predominantly due to nephrotoxicity (13). Even so, it remains in use as a standard chemotherapeutic agent for the treatment of numerous types of cancer, including ovarian, cervical and prostate cancer (1417). A previous study demonstrated that patients treated with DDP in combination with β-elemene were able to better tolerate the chemotherapy, which afforded an improved treatment for hormone-refractory prostate cancer (18). Therefore, how to reduce the toxicity associated with DDP treatment is a keenly studied topic in cancer research.

The present study aimed to provide important insights into the effects of LSD1 knockdown and its interplay with DDP on the proliferation, apoptosis and invasion of PC3 human prostate cancer cells. In addition, the present study revealed whether LSD1 knockdown could increase the sensitivity of DDP for the treatment of prostate cancer. The results may provide important implications for the development of novel therapeutic strategies.

Materials and methods

Cell line and culture

The PC3 human prostate cancer cell line was purchased from the American Type Culture Collection (Manassas, VA, USA). The cells were grown in Gibco™ RPMI-1640 medium (Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS) and 2 mM glutamine (both Sigma-Aldrich, St. Louis, MO, USA) at 37°C in a humidified atmosphere containing 5% CO2. Plasmids encoding LSD1 small interfering (si)RNA or mock vehicle pCMV-G&NR-U6-shRNA (GeneChem Co., Ltd., Shanghai, China) were transfected into the PC3 cells in 6-well plates using a lentiviral vector (JRDUN Biotechnology, Shanghai, China) and Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocols. Three sequences of the LSD1 were used, as follows: Short hairpin (sh)LSD1-1, 5′-ACGAAAGTGTCTCCGTTGA-3′; shLSD1-2, 5′-CCGACATGGCTTTCTCTTT-3′; and shLSD1-3, 5′-TCGACAGTGACCCCTTATA-3′. The cells were split twice weekly, and cells in the logarithmic growth phase were used for subsequent experiments.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from the cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), and the mRNA was reverse transcribed into cDNA using the TIANScript RT kit (Tiangen Biotech. Co. Ltd., Shanghai, China). Subsequently, qPCR was conducted using the SYBR Green PCR Master mix (Thermo Fisher Scientific, Inc.) and the ABI 7300 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.), in which glyceraldehyde-3-phosphate dehydrogenase (GADPH) was used as the reference gene. The following primers were used: LSD1, forward (F) 5′-AAGCAGGAGGACTTCAAGAC-3′, reverse (R) 5′-GCAGTGTGCGGTTTCTAATG-3′; GAPDH, F 5′-CACCCACTCCTCCACCTTTG-3′ and R 5′-CCACCACCCTGTTGCTGTAG-3′ (Generay Biotech Co., Ltd., Shanghai, China). The PCR cycling conditions were as follows: 95°C for 10 min, followed by 40 cycles at 95°C for 15 sec and 60°C for 1 min. RT-qPCR data were analyzed with SDS 2.3 software. Each experiment was repeated three times and the relative mRNA expression levels of LSD1 were calculated using the 2−ΔΔCq method (19).

Cell Counting kit 8 (CCK-8) assay

The viability of the PC3 cells was measured using a CCK-8 kit (Boster Biological Technology, Ltd., Wuhan, China). The cells were seeded into a 96-well microplate at a density of 5×103/well and incubated for 24 h. The peripheral wells of the microplate contained phosphate-buffered saline (PBS) only. The cells were divided into four groups: Mock vehicle group, LSD1 siRNA group, mock vehicle + DDP group or LSD1 siRNA + DDP group, according to the experimental design. The cells were treated with 5 µg/ml DDP (Beyotime Institute of Biotechnology, Haimen, China), after which the cells were incubated for a further 24 h prior to the assay. A total of 10 µl CCK8 solution was added to each well containing PC3 cells, and the cells were incubated for 0, 24, 48 or 72 h. Finally, the absorbance of each well was measured at 490 nm using a Gemini XPS microplate reader (Molecular Devices, LLC, Sunnyvale, CA, USA).

Flow cytometric analysis

The PC3 cells (5×105 cells/ml) were inoculated into 6-well plates. Each group comprised three double wells on the plate. Following a 24 h incubation, the groups were generated by addition of the appropriate reagents to the cells, and the cells were incubated for a further 24 h. The flow cytometric analysis was performed according to the protocol of the Annexin V-Fluorescein Isothiocyanate Apoptosis Detection kit (Abcam, Cambridge, UK). The apoptotic rates were analyzed immediately using a FACSCalibur™ flow cytometer (BD Biosciences, San Jose, CA, USA).

Cell invasive capability measured using a Transwell assay

Following the removal of the culture medium from each group, the cells were digested with trypsin and diluted to 1×105/ml in serum-free Dulbecco's Modified Eagle's Medium containing 1% FBS (GE Healthcare Life Sciences, Logan, UT, USA). A total of 800 µl culture medium containing 10% FBS was added to the coated lower chambers of a Transwell system. Matrigel (BD Biosciences) was coated onto the upper chambers of the Transwell system, after which 200 µl cell suspension was added to the upper chambers at a density of 5×104/well. The plate was cultured for 24 h, after which the cells on the upper layer were removed. The cells that had migrated to the lower layer were washed with PBS, fixed with methanol and stained with 1% crystal violet. The invasive cells were counted in five fields for each sample under an inverted microscope (BX51; Olympus Corporation, Tokyo, Japan), and the results were averaged. The experiments were repeated three times.

Western blot analysis

Total protein was extracted from the cell samples using radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology), and was quantified using the Bicinchoninic Acid Protein Assay kit (Thermo Fisher Scientific, Inc.). Subsequently, equal volumes of protein (30 µg) were separated by 12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis followed by immunoblotting onto a nitrocellulose membrane (EMD Millipore, Billerica, MA, USA) using an electrophoretic transfer cell (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The membranes were blocked with 5% skimmed milk, followed by incubation overnight at 4°C with the following primary antibodies: Monoclonal anti-E-cadherin (#14472; 1:1,000), polyclonal anti-cyclo-oxygenase-2 (COX-2; #4842; 1:1,000), monoclonal anti-Smad2/3 (#8685; 1:1,000), monoclonal anti-phosphorylated (p)-Smad2/3 (#8828; 1:1,000), monoclonal anti-cyclin D (#2978S; 1:1,000), polyclonal anti-p-Akt (#9271; 1:1,000), polyclonal anti-Akt (#9272; 1:1,000), monoclonal anti-extracellular signal-regulated kinase (ERK; #4695; 1:1,000), monoclonal anti-p-ERK (#4376; 1:1,000) and monoclonal anti-GAPDH (#5174; 1:1,500) from Cell Signaling Technology, Inc. (Danvers, MA, USA), and polyclonal anti-transforming growth factor-β1 (TGF-β1; ab92486; 1:800) and anti-vascular endothelial growth factor (VEGF; ab46154; 1:1,000) from Abcam. Subsequently, the blots were washed three times with PBS and incubated with horseradish peroxidase-conjugated goat anti-mouse (A0216; 1:1,000) or goat anti-rabbit (A0208; 1:1,000; both Beyotime Institute of Biotechnology) secondary antibodies for 1 h at room temperature. The bands were detected by reaction with enhanced chemiluminescence detection system reagents (EMD Millipore) and exposure to X-ray film (Kodak, Rochester, NY, USA), which was subsequently developed and used to capture photographic images. GADPH was used to normalize the protein expression. Band intensities were analyzed using ImageJ 1.49 software (https://imagej.nih.gov/ij/).

Statistical analysis

All data are presented as the mean ± standard deviation. The data were evaluated using the Prism 5.0 statistical software package (GraphPad Software Inc., San Diego, CA, USA). The two-tailed Student's t-test was used to evaluate statistical differences between two groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Proliferation of PC3 cells is decreased following LSD1 knock-down and treatment with DDP

The present study hypothesized that LSD1 may have an important role in PC3 cells, which has not been previously investigated. Lentiviral-mediated RNA interference technology was used to establish a stably transfected LSD1 knockdown PC3 cell line, and the CCK-8 colorimetric assay was subsequently used to determine cell proliferation. As shown in Fig. 1A, the mRNA expression levels of LSD1 in the knockdown group were decreased to <60% of the control levels (P<0.01), whereas the mRNA expression levels of LSD1 in the mock group (i.e. control cells which were transfected with an irrelevant interference sequence) exhibited no significant changes. All of the groups contained similar cell numbers at the 0 h time point; however, proliferation of the LSD1 knockdown PC3 cells was significantly decreased after 24, 48 and 72 h, as compared with the mock group (P<0.01; Fig. 1B). Furthermore, compared with the mock group, DDP (at a concentration of 5 µg/ml) exerted a marked inhibitory effect on PC3 cell proliferation. Notably, the LSD1 knockdown + DDP group demonstrated a more marked inhibition on PC3 cell proliferation, as compared with the DDP or LSD1 knockdown groups (P<0.05). These findings indicate that the proliferative capability of the PC3 cells was decreased following LSD1 knockdown, thus suggesting that LSD1 may contribute to the proliferation of PC3 cells, and that siRNA interference may result in reduced cell growth. Furthermore, DDP had similar effects to LSD1 siRNA, and a combination of LSD1 knockdown and DDP treatment produced a synergistic effect.

LSD1 knockdown and DDP treatment inhibit PC3 cell proliferation via regulation of the Akt and ERK signal transduction pathways

The results of the present study revealed that cell proliferation was inhibited when LSD1 was knocked down and the cells were co-treated with DDP, as compared with the mock group. Since the Akt and ERK signal transduction pathways are reported to modulate cell proliferation and tumorigenicity (20,21), the protein expression levels of p-Akt and p-ERK were subsequently investigated. Furthermore, cyclin D has a crucial role in the Akt signal pathway as a regulatory protein of the cell cycle (22), and therefore the protein expression levels of cyclin D were also investigated. The protein expression levels of p-Akt, p-ERK and cyclin D1 were downregulated in the treatment groups (LSD1, mock + DDP, LSD1 + DDP), as compared with the mock group (Fig. 1C and D). These results suggest that LSD1 RNA interference and treatment with DDP may inhibit PC3 cell proliferation via regulation of the Akt and ERK signal pathways.

Percentage of PC3 cells in the early apoptotic phase is increased following LSD1 knockdown and treatment with DDP

In addition to regulating PC3 cell growth, LSD1 may be involved in regulating cell apoptosis. As shown in Fig. 2A and B the proportion of PC3 cells in the early apoptotic phase was significantly increased following LSD1 knockdown, as compared with the mock group. Furthermore, it was revealed that DDP contributes to the increased levels of apoptosis in the PC3 cells, and that treatment with DDP exerted a synergistic action on PC3 cell apoptosis when combined with LSD1 siRNA. These results suggest that a combination of LSD1 knockdown and DDP treatment may contribute to the increased percentage of PC3 cells in the early apoptotic stage.

Invasive capability of PC3 cells is decreased following LSD1 knockdown and treatment with DDP via regulation of the TGF-β1/Smad2/3 signal transduction pathway

Invasive capability is essential for the malignant progression of tumors. As shown in Fig. 3A and B, the invasive capability of the PC3 cells was decreased following LSD1 knockdown or treatment with DDP, as compared with the mock group. Furthermore, a combination of LSD1 knockdown and DDP treatment exerted a synergistic effect on the decline in invasive ability. These results suggest that LSD1 may have a crucial role in the invasion of PC3 cells.

TGF-β1 is a cytokine peptide, which is associated with various biological roles in cancer (23). The Smad proteins act as substrates of the TGF-β1 receptor, and their activation by phosphorylation propagates the TGF-β1 signal transduction. In particular, the TGF-β1/Smad2/3 signaling pathway is involved in mechanisms underlying the invasion and metastasis of prostate cancer cells, a process which is also regulated by tumor angiogenesis, the host immune system, the cells themselves and the surrounding matrix microenvironment (2426). Consequently, the protein expression levels of TGF-β1, p-Smad2/3, VEGF, COX-2 and E-cadherin, all of which are associated with the TGF-β1/Smad2/3 signal transduction pathway, were investigated. The results revealed that the expression levels of TGF-β1, p-Smad2/3, VEGF and COX-2 in the DDP-treated cells were downregulated, whereas the protein expression levels of E-cadherin were upregulated. Furthermore, the combination of LSD1 knockdown and treatment with DDP exerted a synergistic effect, as compared with either treatment taken in isolation or with the mock cells (Fig. 3C–E).

Discussion

Prostate cancer is a complex disease, and numerous controversies are associated with aspects of different treatment strategies (27). The identification of genetic and molecular events that may improve the early detection of prostate cancer, or that could be used as therapeutic targets, is a top priority in this line of study.

LSD1 is a flavin-dependent amine oxidase, which has been reported to interact with the AR (9). Previous studies have reported that LSD1 is overexpressed in prostate cancer (9,28). DDP is an inorganic compound that is widely used in cancer therapy (12). In the present study, the antiproliferative, proapoptotic and anti-invasive effects of LSD1 knockdown and DDP treatment, either in isolation or in combination, on PC3 prostate cancer cells were investigated. The results of the present study may lead to an improved understanding regarding how LSD1 knockdown and DDP treatment affect the physiological activity of prostate cancer cells, thereby providing a novel target for therapeutic intervention in prostate cancer.

A combination of LSD1 knockdown and DDP treatment effectively suppressed the proliferation of PC3 cells. The Akt and ERK signaling pathways have been reported to be important pathways closely associated with cell proliferation (29,30). In the present study, western blotting results suggested that the protein expression levels of the associated proteins, p-Akt, cyclin D1 and p-ERK, were markedly decreased in the LSD1 siRNA, DDP treatment and LSD1 siRNA + DDP cell groups, as compared with the mock group. These findings indicated that LSD1 knockdown and DDP treatment may effectively inhibit the proliferation of PC3 cells via regulation of the Akt and ERK signaling pathways. Flow cytometry was also performed to determine the extent of apoptosis in the prostate cancer cells. All of the treatments, i.e. LSD1 knockdown and treatment with DDP, either alone or in combination, resulted in an increased induction of PC3 cell apoptosis.

Tumor cell invasion and metastasis are complicated by the existence, and interplay, of several mechanisms (31). The effects of LSD1 knockdown on the invasive capability of the PC3 prostate cancer cells were examined in the present study. The results demonstrated that knockdown of LSD1, in combination with DDP, suppressed the invasion of prostate cancer cells. TGF-β is a cytokine peptide, which exerts various biological activities. A previous study demonstrated that TGF-β1 was able to promote the invasion and metastasis of prostate tumor cells (32). Smad proteins are the sole substrates of TGF-β1 identified in the TGF-β1 signal transduction pathway (33). In the present study, the protein expression levels of TGF-β1 and p-Smad2/3 were decreased, which implied that this signaling pathway may be primarily responsible for the process by which LSD1 knockdown and DDP treatment inhibited the invasion of PC3 cells. TGF-β1/Smad2/3 signaling is known to promote specific mechanisms underlying the processes of cell invasion and metastasis in prostate cancer, via regulation of angiogenesis, the immune defense system, changes in the substrate microenvironment or alterations to the cells themselves (3436).

The formation of blood vessels is crucial in tumor growth, invasion and metastasis (37). The present study revealed that VEGF, as one of the most important promoters of angiogenesis (38), was downregulated in the LSD1 siRNA group. In addition, epithelial-mesenchymal transition is considered to be a key step in the process of tumor metastasis. For example, E-cadherin is as a Ca2+-dependent glycoprotein, which performs an essential role in cell invasion and migration (39,40). The results of the present study implied that LSD1 knockdown and DDP treatment inhibited the invasion of the PC3 cells by upregulating the expression levels of E-cadherin. COX-2, which is a rate-limiting enzyme in the prostaglandin biosynthesis pathway, is able to promote tumor angiogenesis (41). The western blotting results of the present study demonstrated that the protein expression levels of COX-2 were decreased in DDP-treated LSD1 knockdown cells, with a similar result obtained for VEGF. These results may verify the hypothesis that LSD1 knockdown inhibits the invasion of PC3 cells by regulating the TGF-β1/Smad2/3 signal pathway, and a combination of LSD1 siRNA and DDP treatment may lead to more pronounced effects.

In conclusion, the present study aimed to identify the proapoptotic, antiproliferative and anti-invasive effects of LSD1 knockdown, in combination with DDP treatment, on PC3 human prostate cancer cells, and to offer an explanation for the underlying mechanism. The results of the present study may provide novel insights into the molecular mechanism underlying the progression and pathogenesis of prostate cancer, and may be useful for the optimization of therapeutic interventions for the treatment of this disease.

References

1 

Center MM, Jemal A, Lortet-Tieulent J, Ward E, Ferlay J, Brawley O and Bray F: International variation in prostate cancer incidence and mortality rates. Eur Urol. 61:1079–1092. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Bechis SK, Carroll PR and Cooperberg MR: Impact of age at diagnosis on prostate cancer treatment and survival. J Clin Oncol. 29:235–241. 2011. View Article : Google Scholar :

3 

Chen CD, Welsbie DS, Tran C, Baek SH, Chen R, Vessella R, Rosenfeld MG and Sawyers CL: Molecular determinants of resistance to antiandrogen therapy. Nat Med. 10:33–39. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Shahinian VB, Kuo YF, Freeman JL and Goodwin JS: Risk of fracture after androgen deprivation for prostate cancer. N Engl J Med. 352:154–164. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Lv T, Yuan D, Miao X, Lv Y, Zhan P, Shen X and Song Y: Overexpression of LSD1 promotes proliferation, migration and invasion in non-small cell lung cancer. PloS One. 7:e350652012. View Article : Google Scholar

6 

Zhao ZK, Dong P, Gu J, Chen L, Zhuang M, Lu WJ, Wang DR and Liu YB: Overexpression of LSD1 in hepatocellular carcinoma: A latent target for the diagnosis and therapy of hepatoma. Tumor Biol. 34:173–180. 2013. View Article : Google Scholar

7 

Qin Y, Zhu W, Xu W, Zhang B, Shi S, Ji S, Liu J, Long J, Liu C, Liu L, et al: LSD1 sustains pancreatic cancer growth via maintaining HIF1α-dependent glycolytic process. Cancer Lett. 347:225–232. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Kauffman EC, Robinson BD, Downes MJ, Powell LG, Lee MM, Scherr DS, Gudas LJ and Mongan NP: Role of androgen receptor and associated lysine-demethylase coregulators, LSD1 and JMJD2A, in localized and advanced human bladder cancer. Mol Carcinog. 50:931–944. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Metzger E, Wissmann M, Yin N, Müller JM, Schneider R, Peters AH, Günther T, Buettner R and Schüle R: LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature. 437:436–439. 2005.PubMed/NCBI

10 

Kahl P, Gullotti L, Heukamp LC, Wolf S, Friedrichs N, Vorreuther R, Solleder G, Bastian PJ, Ellinger J, Metzger E, et al: Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence. Cancer Res. 66:11341–11347. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Seligson DB, Horvath S, Shi T, Yu H, Tze S, Grunstein M and Kurdistani SK: Global histone modification patterns predict risk of prostate cancer recurrence. Nature. 435:1262–1266. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Natile G and Coluccia M: Current status of trans-platinum compounds in cancer therapy. Coord Chem Rev. 216:383–410. 2001. View Article : Google Scholar

13 

Miller RP, Tadagavadi RK, Ramesh G and Reeves WB: Mechanisms of Cisplatin nephrotoxicity. Toxins (Basel). 2:2490–2518. 2010. View Article : Google Scholar

14 

Florea AM and Büsselberg D: Cisplatin as an anti-tumor drug: Cellular mechanisms of activity, drug resistance and induced side effects. Cancers (Basel). 3:1351–1371. 2011. View Article : Google Scholar

15 

Gumulec J, Balvan J, Sztalmachova M, Raudenska M, Dvorakova V, Knopfova L, Polanska H, Hudcova K, Ruttkay-Nedecky B, Babula P, et al: Cisplatin-resistant prostate cancer model: Differences in antioxidant system, apoptosis and cell cycle. Int J Oncol. 44:923–933. 2014.

16 

Rose PG, Sill MW, McMeekin DS, Ahmed A, Salani R, Yamada SD, Wolfson AH, Fusco N and Fracasso PM: A phase I study of concurrent weekly topotecan and cisplatin chemotherapy with whole pelvic radiation therapy in locally advanced cervical cancer: A gynecologic oncology group study. Gynecol Oncol. 125:158–162. 2012. View Article : Google Scholar

17 

Stordal B, Hamon M, McEneaney V, Roche S, Gillet JP, O'Leary JJ, Gottesman M and Clynes M: Resistance to paclitaxel in a cisplatin-resistant ovarian cancer cell line is mediated by P-glycoprotein. PloS One. 7:e407172012. View Article : Google Scholar : PubMed/NCBI

18 

Li QQ, Wang G, Reed E, Huang L and Cuff CF: Evaluation of cisplatin in combination with β-elemene as a regimen for prostate cancer chemotherapy. Basic Clin Pharmacol Toxicol. 107:868–876. 2010.PubMed/NCBI

19 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar

20 

Jin F, Irshad S, Yu W, Belakavadi M, Chekmareva M, Ittmann MM, Abate-Shen C and Fondell JD: ERK and AKT signaling drive MED1 overexpression in prostate cancer in association with elevated proliferation and tumorigenicity. Mol Cancer Res. 11:736–747. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Rick FG, Schally AV, Szalontay L, Block NL, Szepeshazi K, Nadji M, Zarandi M, Hohla F, Buchholz S and Seitz S: Antagonists of growth hormone-releasing hormone inhibit growth of androgen-independent prostate cancer through inactivation of ERK and Akt kinases. Proc Natl Acad Sci USA. 109:1655–1660. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Harashima N, Inao T, Imamura R, Okano S, Suda T and Harada M: Roles of the PI3K/Akt pathway and autophagy in TLR3 signaling-induced apoptosis and growth arrest of human prostate cancer cells. Cancer Immunol Immunother. 61:667–676. 2012. View Article : Google Scholar

23 

Leivonen SK and Kähäri VM: Transforming growth factor-beta signaling in cancer invasion and metastasis. Int J Cancer. 121:2119–2124. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Nicholson B and Theodorescu D: Angiogenesis and prostate cancer tumor growth. J Cell Biochem. 91:125–150. 2004. View Article : Google Scholar

25 

Wikström P, Damber J and Bergh A: Role of transforming growth factor-beta1 in prostate cancer. Microsc Res Tech. 52:411–419. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2:442–454. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Denmeade SR and Isaacs JT: A history of prostate cancer treatment. Nat Rev Cancer. 2:389–396. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Kashyap V, Ahmad S, Nilsson EM, Helczynski L, Kenna S, Persson JL, Gudas LJ and Mongan NP: The lysine specific demethylase-1 (LSD1/KDM1A) regulates VEGF-A expression in prostate cancer. Mol Oncol. 7:555–566. 2013. View Article : Google Scholar : PubMed/NCBI

29 

De Luca A, Maiello MR, D'Alessio A, Pergameno M and Normanno N: The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: Role in cancer pathogenesis and implications for therapeutic approaches. Expert Opin Ther Targets. 16(Suppl 2): S17–S27. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Kinkade CW, Castillo-Martin M, Puzio-Kuter A, Yan J, Foster TH, Gao H, Sun Y, Ouyang X, Gerald WL, Cordon-Cardo C and Abate-Shen C: Targeting AKT/mTOR and ERK MAPK signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model. J Clin Invest. 118:3051–3064. 2008.PubMed/NCBI

31 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Danielpour D: Functions and regulation of transforming growth factor-beta (TGF-beta) in the prostate. Eur J Cancer. 41:846–857. 2005. View Article : Google Scholar : PubMed/NCBI

33 

ten Dijke P and Hill CS: New insights into TGF-beta-Smad signalling. Trends Biochem Sci. 29:265–273. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Tuxhorn JA, McAlhany SJ, Yang F, Dang TD and Rowley DR: Inhibition of transforming growth factor-beta activity decreases angiogenesis in a human prostate cancer-reactive stroma xenograft model. Cancer Res. 62:6021–6025. 2002.PubMed/NCBI

35 

Donkor MK, Sarkar A, Savage PA, Franklin RA, Johnson LK, Jungbluth AA, Allison JP and Li MO: T cell surveillance of oncogene-induced prostate cancer is impeded by T cell-derived TGF-β1 cytokine. Immunity. 35:123–134. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Micalizzi DS, Farabaugh SM and Ford HL: Epithelial-mesenchymal transition in cancer: Parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia. 15:117–134. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Kirsch M, Schackert G and Black PM: Angiogenesis, metastasis, and endogenous inhibition. J Neurooncol. 50:173–180. 2000. View Article : Google Scholar

38 

Ferrara N, Gerber HP and LeCouter J: The biology of VEGF and its receptors. Nat Med. 9:669–676. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Pàmies P: E-cadherin-guided migration. Nat Mater. 13:6642014. View Article : Google Scholar

40 

Canel M, Serrels A, Frame MC and Brunton VG: E-cadherin-integrin crosstalk in cancer invasion and metastasis. J Cell Sci. 126:393–401. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Muraki C, Ohga N, Hida Y, Nishihara H, Kato Y, Tsuchiya K, Matsuda K, Totsuka Y, Shindoh M and Hida K: Cyclooxygenase-2 inhibition causes antiangiogenic effects on tumor endothelial and vascular progenitor cells. Int J Cancer. 130:59–70. 2012. View Article : Google Scholar

Related Articles

Journal Cover

September-2016
Volume 14 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen ZY, Chen H, Qiu T, Weng XD, Guo J, Wang L and Liu XH: Effects of cisplatin on the LSD1-mediated invasion and metastasis of prostate cancer cells Retraction in /10.3892/mmr.2023.13027. Mol Med Rep 14: 2511-2517, 2016
APA
Chen, Z., Chen, H., Qiu, T., Weng, X., Guo, J., Wang, L., & Liu, X. (2016). Effects of cisplatin on the LSD1-mediated invasion and metastasis of prostate cancer cells Retraction in /10.3892/mmr.2023.13027. Molecular Medicine Reports, 14, 2511-2517. https://doi.org/10.3892/mmr.2016.5571
MLA
Chen, Z., Chen, H., Qiu, T., Weng, X., Guo, J., Wang, L., Liu, X."Effects of cisplatin on the LSD1-mediated invasion and metastasis of prostate cancer cells Retraction in /10.3892/mmr.2023.13027". Molecular Medicine Reports 14.3 (2016): 2511-2517.
Chicago
Chen, Z., Chen, H., Qiu, T., Weng, X., Guo, J., Wang, L., Liu, X."Effects of cisplatin on the LSD1-mediated invasion and metastasis of prostate cancer cells Retraction in /10.3892/mmr.2023.13027". Molecular Medicine Reports 14, no. 3 (2016): 2511-2517. https://doi.org/10.3892/mmr.2016.5571