G2/M accumulation in prostate cancer cell line PC-3 is induced by Cdc25 inhibitor 7-chloro-6-(2-morpholin-4-ylethylamino) quinoline-5, 8-dione (DA 3003-2)

  • Authors:
    • Kaoru Nemoto
  • View Affiliations

  • Published online on: July 1, 2010     https://doi.org/10.3892/etm_00000101
  • Pages: 647-650
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cdc25 phosphatases are dual-specific phosphatases that play a role in cell cycle progression. In many human cancers, Cdc25 phosphatases are overexpressed as compared with normal tissues. In addition, overexpression of Cdc25 phosphatases in prostate cancer is correlated with disease progression. The antiproliferative efficacy of Cdc25 phosphatase inhibitor 7-chloro-6-(2-morpholin-4-ylethylamino) quinoline-5, 8-dione (DA 3003-2) was investigated in the PC-3 asynchronous human prostate cancer cell line using a cell-based assay. The time course changes in cell cycle distribution and the modulation of cell cycle regulators after DA 3003-2 administration were analyzed using the MTT assay. We found that the relative IC50 of DA 3003-2 was 2-fold lower as compared with its congener (2-mercaptoethanol)-3-methyl-1, 4-naphthoquinone (NSC 672121). Asynchronous PC-3 cells accumulated in the G2/M phase at 24 h after treatment with 10 µM DA 3003-2 or 20 µM NSC 672121, which represent IC70 concentrations. Treatment of cells with DA 3003-2 caused hyperphosphorylation of Cdc2 tyr15 in cyclin B1 and cyclin A complexes. DA 3003-2 did not downregulate the protein expression levels of Cdc25s, cyclins and cyclin-dependent kinases (Cdks). To conclude, after DA 3003-2 administration asynchronous PC-3 cells accumulated in the G2/M phase, with hyperphosphorylation of the G2/M cyclin-Cdk complex.

Introduction

Cell cycle progression and cell proliferation are controlled in part by a balance between cyclins, cyclin-dependent kinases (Cdks) and phosphatases. Various types of human cancers are characterized by uncontrolled cell growth which leads to poor patient prognosis. One of the mechanisms involved in uncontrolled cell growth is thought to be the change in basic cell cycle regulation caused by cancer-associated mutations, overexpression of cell cycle-regulated protein, as well as by the loss of Cdk inhibitor expression. Cdc25 is a dual-specificity protein tyrosine phosphatase which catalyzes the dephosphorylation and activation of cyclin-Cdk complexes through the removal of inhibitory phosphates. The Cdc25 phosphatase family comprises three related gene products, namely Cdc25A, Cdc25B and Cdc25C. Cdc25A and Cdc25B cooperate with activated Ras to induce oncogenic focus formation in rat embryonic fibroblasts. Moreover, overexpression of Cdc25 phosphatases in human cancers correlates with disease progression and is an indicator of poor patient prognosis (1,2). Taken together, these results suggest that Cdc25 phosphatases have potential as a potent small molecular target for cancer therapy.

In prostate cancer, Cdc25A, Cdc25B and Cdc25C are upregulated in cancerous lesions relative to non-cancerous lesions, and levels are increased in higher Gleason grade tumors (35). A number of novel Cdc25 inhibitors from the compounds library at the University of Pittsburgh have been screened with a focus on compound structures, such as quinine or naphthoquinone structures (68). In the present study, a more selective naphthoquinone Cdc25 inhibitor, 7-chloro-6-(2-morpholin-4-ylethylamino) quinoline-5, 8-dione (DA 3003-2), was generated from the drug library at the University of Pittsburgh, and its molecular mechanisms of action were investigated using the PC-3 human prostate cancer cell line.

Materials and methods

Cell culture, chemicals and antibodies

PC-3 human prostate cancer cells were obtained from the American Type Culture Collection (Bethesda, MD). The cells were cultured at 37°C in RPMI-1640 medium supplemented with 10% fetal bovine serum in a humidified atmosphere of 5% CO2. DA 3003-2 and (2-mercaptoethanol)-3-methyl-1, 4-naphthoquinone (NSC 672121) were generated as described previously (7). These compounds were solubilized so that the final concentration of dimethyl sulfoxide (DMSO) was <0.1% when added to the cells. The following antibodies were used: anti-Cdc25A (Ab3) purchased from NeoMarkers, Inc. (Fremont, CA); anti-Cdc25B from Transduction Laboratories (Lexington, KY); and anti-Cdc25C (C-2), anti-Cdk2 (D-12), anti-Cdc2 (17), anti-phospho-Cdc2/Cdk2 (Tyr15), anti-cyclin A (H-432), anti-cyclin B1 (GNS1) and anti-actin (C-2) from Santa Cruz Biotechnology (Santa Cruz, CA).

MTT assay

The sensitivity of the cells to NSC 672121 and DA 3003-2 was determined by a microtiter assay. Cells (4×103) were plated in 96-well microtiter plates, cultured for 24 h and exposed continuously to 0.3–30 μM of NSC 672121 or DA 3003-2 for 48 h. The viability of the cells was assayed by determining the color development resulting from the reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) spectrophotometrically at 540 nm, as described previously (7).

Flow cytometry

PC-3 cells were plated at 5×105 cells/dish and maintained for 24 h. Cells were treated with DMSO, NSC 672121 or DA 3003-2. After 24 h, cells were trypsinized and washed with phosphate-buffered saline (PBS). The harvested cells were stained with a solution containing 50 μg/ml propidium iodide (PI) and 250 μg/ml RNase A. Flow cytometric analysis was conducted using EPICS XL™ and XL-MCL™ systems (Beckman Coulter, Inc.).

Western blotting and immunoprecipitation

Vehicle- or compound-treated cells were harvested by scraping and were resuspended in lysis buffer (30 mM HEPES, 1% Triton X-100, 10 mM NaCl, 10% glycerol, 5 mM MgCl2, 25 mM NaF, 1 mM EDTA and 0.2 mM Na3VO4, pH 7.6) with protease inhibitors [10 μg/ml leupeptin, 10 μg/ml aprotinin and 100 μg/ml 4-(2-aminoethyl)benzenesulfonyl fluoride]. The samples were briefly vortexed and centrifuged at 13,000 x g for 30 min. The total protein concentration was determined using the Bradford protein assay (BioRad, CA). To immunoprecipitate cyclin B1, cyclin A, Cdc2 and Cdk2 proteins, 2 μg of anti-cyclin B1, anti-cyclin A, anti-Cdc2 or anti-Cdk2 antibodies and Protein G Sepharose 4B were incubated with 1 mg of lysates for 16 h. Beads were washed three times by vortexing with lysis buffer at each step. Equal amounts of protein or supernatant were resolved by SDS-PAGE and transferred to a nitrocellulose membrane. A chemiluminescence detection system (Western Lightning™; Perkin Elmer Life Sciences, Boston, MA) was used for immunocomplex detection.

Results

Cdc25 inhibitor induced G2/M accumulation in asynchronous PC-3 cells

To determine the cytotoxicity of DA 3003-2 in PC-3 cells as compared with the cytotoxic effects of the non-specific Cdc25 inhibitor NSC 672121 [2-(2-mercaptoethanol)-3-methyl-1, 4-naphthoquinone], the MTT assay was performed. The IC50 of DA 3003-2 in PC-3 cells was 2-fold higher than that of NSC 672121, and the IC50 values of NSC 672121 and DA 3003-2 were ∼10 and 5 μM, respectively (Fig. 1). Tamura et al reported that NSC 672121 induced dual G1 and G2/M arrest in synchronized tsFT210 cells (9). Next, the changes in cell cycle distribution induced in asynchronous PC-3 cells by NSC 672121 and DA 3003-2 were analyzed. After 24 h, PC-3 cells had accumulated in the G2/M phase in a concentration-dependent manner as a result of the administration of both compounds (Fig. 2, Table I). DA 3003-2 induced G2/M accumulation at a concentration that was lower than that for NSC 672121 supporting the results of the MTT assay.

Table I.

Cell cycle distribution 24 h after treatment with and without Cdc25 inhibitors.

Table I.

Cell cycle distribution 24 h after treatment with and without Cdc25 inhibitors.

G1SG2/M
No treatment35.8±2.48.2±0.452.5±2.8
DMSO35.7±1.58.0±1.153.2±1.0
DA 3003-2
  5 μM33.9±2.312.3±1.546.8±1.6
  10 μM19.6±1.87.3±0.670.3±1.4
NSC 672121
  10 μM36.3±1.07.9±0.654.9±1.8
  20 μM25.7±2.05.5±0.767.2±1.3
Hyperphosphorylation of Cdc2 Tyr15 in the cyclin-Cdk complex by DA 3003-2

Dephosphorylation of Cdk tyrosine by Cdc25s is necessary for the full activation of the cyclin-Cdk complex. NSC 672121 and 6-chloro-7-(2-morpholin-4-ylethylamino)quinoline-5, 8-dione (NSC 663284), which is a regioisomer of DA 3003-2, were previously found to increase total Cdc2 Tyr15 levels in parallel with the inhibition of Cdc2 kinase activity. In the present study, whether DA 3003-2 and NSC 672121 affect the Tyr15 phosphorylation status of Cdc2 in cyclin-Cdc2 complexes was investigated using a cell-based assay. Cyclin B1 or cyclin A were immunoprecipitated from whole cell lysates harvested 1 h after exposure to 5 or 10 μM DA 3003-2 or 20 μM NSC 672121. The lysate proteins were separated by SDS-PAGE and immunoblotted with antibodies to both Cdc2 phospho-Tyr15 and Cdc2. As shown in Fig. 3, 10 μM DA 3003-2 and 20 μM NSC 672121 hyperphosphorylated Cdc2 Tyr15 in both cyclin-Cdc2 complexes within 1 h. Cdc25 is an unstable protein and is easily degraded by the proteasome pathway. Therefore, the modulation of cell cycle regulator expression levels was investigated using Western blotting. As shown in Fig. 4, Cdc25, G2/M cyclins and Cdc2 levels were not downregulated by DA 3003-2 administration.

Discussion

DA 3003-2 obtained from the compounds library at the University of Pittsburgh was screened using an in vitro phosphatase assay and was previously found to be more selective against Cdc25B2 relative to dual phosphatase VHR or protein tyrosine phosphatase PTP1B (6). It has already been revealed that NSC 663284, which is a regioisomer of DA 3003-2, inhibited Cdc25 activity by binding directly to the Cdc25 catalytic domain (10). On the other hand, Brisson et al suggested that the detection of differences in the cell cycle profile of asynchronous cells using Cdc25 inhibitors was difficult (11). In the present study, it was revealed that G2/M accumulation was induced in asynchronous cells using the Cdc25 inhibitor DA 3003-2 due to the hyperphosphorylation of the G2/M cyclin-Cdk complex. A Cdc25 overexpression study will be required to fully clarify the relationship between the inhibition of Cdc25 activity and G2/M accumulation using a cell-based assay. Moreover, as many different pathways feed into cell cycle regulation, non-Cdc25-specific cellular insults are also expected to cause G2/M arrest. However, the results of the present study confirm the potency of the Cdc25 inhibitor.

In both early and advanced stage prostate cancer etiology, the androgen receptor plays an important role (12,13). Androgen ablation remains the primary course of treatment for all patients with metastatic disease. These therapies are initially effective. However, recurrent tumors arise within a median time of 2–3 years. The balance of androgen receptor co-regulators is associated with the androgen refractory mechanism. Cdc25B directly acts as the co-activator of the androgen receptor and, in contrast, Cdc25A acts as the co-repressor (3,4). Unfortunately, it is difficult to use previously screened Cdc25 inhibitors as molecular targeting agents for androgen co-regulators in androgen refractory prostate cancer. This is due to the fact that the actions of Cdc25s related to androgen receptors have nothing to do with Cdc25 phosphatase activity. On the other hand, several investigators have shown increased expression of cyclin B1 in human prostate cancer (1416). In addition, Maddison et al demonstrated increased cyclin B1 in poorly differentiated and androgen refractory cancers in the TRAMP mouse model of prostate cancer (16). The levels of Cdc25C, which is an activator of the Cdc2/cyclin B complex, increased in prostate cancer and decreased after anti-androgen therapy. Taken together these results suggest that G2/M transition activators, including Cdc25, are one of the important small molecular targets for androgen refractory prostate cancer treatment. The clinical application of the Cdc25 inhibitor as an anti-cancer drug is expected in the near future.

Acknowledgements

The thoughtful com ments of Professor John S. Lazo (Department of Pharmacology, University of Pittsburgh, Pittsburgh, PA) are sincerely appreciated. I would also like to thank Professor Peter Wipf (Department of Chemistry, University of Pitsburgh, Pittsburgh, PA) for providing the compounds used in the present study.

References

1. 

Kristjánsdóttir K and Rudolph J: Cdc25 phosphatases and cancer. Chem Biol. 11:1043–1045. 2004.

2. 

Boutros R, Lobjois V and Ducommun B: CDC25 phosphatases in cancer cells: key players? Good targets? Nat Rev Cancer. 7:495–507. 2007. View Article : Google Scholar : PubMed/NCBI

3. 

Chiu YT, Han HY, Leung SC, Yuen HF, Chau CW, Guo Z, Qiu Y, Chan KW, Wang X, Wong YC and Ling MT: CDC25A functions as a novel AR corepressor in prostate cancer cells. J Mol Biol. 385:446–456. 2009. View Article : Google Scholar : PubMed/NCBI

4. 

Ngan ES, Hashimoto Y, Ma ZQ, Tsai MJ and Tsai SY: Overexpression of Cdc25B, an androgen receptor coactivator, in prostate cancer. Oncogene. 22:734–739. 2003. View Article : Google Scholar : PubMed/NCBI

5. 

Ozen M and Ittmann M: Increased expression and activity of CDC25C phosphatase and an alternatively spliced variant in prostate cancer. Clin Cancer Res. 11:4701–4706. 2005. View Article : Google Scholar : PubMed/NCBI

6. 

Lazo JS, Aslan DC, Southwick EC, Cooley KA, Ducruet AP, Joo B, Vogt A and Wipf P: Discovery and biological evaluation of a new family of potent inhibitors of the dual specificity protein phosphatase Cdc25. J Med Chem. 44:4042–4049. 2001. View Article : Google Scholar : PubMed/NCBI

7. 

Lazo JS, Nemoto K, Pestell KE, Cooley K, Southwick EC, Mitchell DA, Furey W, Gussio R, Zaharevitz DW, Joo B and Wipf P: Identification of a potent and selective pharmacophore for Cdc25 dual specificity phosphatase inhibitors. Mol Pharmacol. 61:720–728. 2002. View Article : Google Scholar : PubMed/NCBI

8. 

Lazo JS and Wipf P: Is Cdc25 a druggable target? Anticancer Agents Med Chem. 8:837–842. 2008. View Article : Google Scholar : PubMed/NCBI

9. 

Tamura K, Southwick EC, Kerns J, Rosi K, Carr BI, Wilcox C and Lazo JS: Cdc25 inhibition and cell cycle arrest by a synthetic thioalkyl vitamin K analogue. Cancer Res. 60:1317–1325. 2000.PubMed/NCBI

10. 

Pu L, Amoscato AA, Bier ME and Lazo JS: Dual G1 and G2 phase inhibition by a novel, selective Cdc25 inhibitor 6-chloro-7 -[corrected] (2-morpholin-4-ylethylamino)-quinoline-5, 8-dione. J Biol Chem. 277:46877–46885. 2002.PubMed/NCBI

11. 

Brisson M, Nguyen T, Vogt A, Yalowich J, Giorgianni A, Tobi D, Bahar I, Stephenson CR, Wipf P and Lazo JS: Discovery and characterization of novel small molecule inhibitors of human Cdc25B dual specificity phosphatase. Mol Pharmacol. 66:824–833. 2004. View Article : Google Scholar : PubMed/NCBI

12. 

Chmelar R, Buchanan G, Need EF, Tilley W and Greenberg NM: Androgen receptor coregulators and their involvement in the development and progression of prostate cancer. Int J Cancer. 120:719–733. 2007. View Article : Google Scholar : PubMed/NCBI

13. 

Balk SP and Knudsen KE: AR, the cell cycle, and prostate cancer. Nucl Recept Signal. 6:1–12. 2008.PubMed/NCBI

14. 

Mashal RD, Lester S, Corless C, Richie JP, Chandra R, Propert KJ and Dutta A: Expression of cell cycle-regulated proteins in prostate cancer. Cancer Res. 56:4159–4163. 1996.PubMed/NCBI

15. 

Kallakury BV, Sheehan CE, Ambros RA, Fisher HA, Kaufman RP Jr, Muraca PJ and Ross JS: Correlation of p34cdc2 cyclin-dependent kinase overexpression, CD44 downregulation, and HER-2/neu oncogene amplification with recurrence in prostatic adenocarcinomas. J Clin Oncol. 16:1302–1309. 1998.PubMed/NCBI

16. 

Maddison LA, Huss WJ, Barrios RM and Greenberg NM: Differential expression of cell cycle regulatory molecules and evidence for a ‘cyclin switch’ during progression of prostate cancer. Prostate. 58:335–344. 2004.

Related Articles

Journal Cover

July-August 2010
Volume 1 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Nemoto K: G2/M accumulation in prostate cancer cell line PC-3 is induced by Cdc25 inhibitor 7-chloro-6-(2-morpholin-4-ylethylamino) quinoline-5, 8-dione (DA 3003-2) . Exp Ther Med 1: 647-650, 2010
APA
Nemoto, K. (2010). G2/M accumulation in prostate cancer cell line PC-3 is induced by Cdc25 inhibitor 7-chloro-6-(2-morpholin-4-ylethylamino) quinoline-5, 8-dione (DA 3003-2) . Experimental and Therapeutic Medicine, 1, 647-650. https://doi.org/10.3892/etm_00000101
MLA
Nemoto, K."G2/M accumulation in prostate cancer cell line PC-3 is induced by Cdc25 inhibitor 7-chloro-6-(2-morpholin-4-ylethylamino) quinoline-5, 8-dione (DA 3003-2) ". Experimental and Therapeutic Medicine 1.4 (2010): 647-650.
Chicago
Nemoto, K."G2/M accumulation in prostate cancer cell line PC-3 is induced by Cdc25 inhibitor 7-chloro-6-(2-morpholin-4-ylethylamino) quinoline-5, 8-dione (DA 3003-2) ". Experimental and Therapeutic Medicine 1, no. 4 (2010): 647-650. https://doi.org/10.3892/etm_00000101