The combinatory effects of PPAR-γ agonist and survivin inhibition on the cancer stem-like phenotype and cell proliferation in bladder cancer cells

  • Authors:
    • Yang Wang
    • Hailin Tan
    • Dongxu Xu
    • Aihui Ma
    • Li Zhang
    • Jiabin Sun
    • Zhaojuan Yang
    • Yongzhong Liu
    • Guowei Shi
  • View Affiliations

  • Published online on: May 8, 2014     https://doi.org/10.3892/ijmm.2014.1774
  • Pages: 262-268
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Strategies for peroxisome proliferator-activated receptor (PPAR) activation or survivin inhibition have potential for cancer therapy. However, whether the combination of these two approaches can be developed as a rational regimen with enhanced efficiency in the inhibition of tumor cells remains to be determined. In this study, the combinatory effect of PPAR-γ agonist and survivin inhibition on bladder cancer cells was investigated. T24 and 5637 cells were treated with 15d-PGJ2 to determine whether 15d-PGJ2 had an inhibitory effect. Cell viability and proliferation were analyzed and efficiency of survivin siRNAs was assessed using western blot analysis. The results showed that, in the human bladder cancer cell lines T24 and 5637, the natural PPAR-γ ligand 15d-PGJ2 significantly decreased cell proliferation and loci formation. The increase in the proportion of apoptotic cells was observed in the cells 48 h after 15d-PGJ2 treatment. Furthermore, 15d-PGJ2 substantially inhibited the levels of stemness-related genes in these cells. The ability of sphere formation was markedly suppressed in the cells treated with 15d-PGJ2. More importantly, the downregulation of survivin with siRNAs significantly enhanced the 15d-PGJ2-mediated induction of cell apoptosis and inhibition of sphere formation. Accordingly, we also found that survivin inhibition significantly enhanced 15d-PGJ2-induced production of reactive oxygen species (ROS) in bladder cancer cells. Taken together, these findings suggest that the combination of 15d-PGJ2 and survivin inhibition play a potentially role in the therapeutical manipulation of bladder cancer.

Introduction

The incidence of bladder cancer, a common urologic cancer, continues to increase annually, ranking as the ninth most common malignancy wordwide (1). Although treatment with systemic chemotherapy is recommended, the prognosis for patients with metastatic bladder cancer is poor. Therefore, improvement of existing therapies and development of alternative therapeutic approaches is critical. Recent advances in the study on tumor-initiating cells, a small subpopulation of tumor cells that contribute to tumor initiation, metastasis and drug-resistance (2), suggest that targeting these cells may lead to novel therapies that can be utilized in the reduction of risk of tumor recurrence.

The peroxisome proliferator-activated receptor-γ (PPAR-γ) is a member of the nuclear receptor superfamily that is activated by its ligands. The activation of PPAR-γ may lead to cell growth arrest, apoptosis, decrease of cell adhesion and migration, and particularly, result in the differentiation of cancer cells (3). The property of their antigrowth and prodifferentiation renders natural and synthetic ligands of PPAR-γ as attractive substances in cancer prevention and treatment (36). However, given that PPAR-γ ligands often trigger crosstalk with other signalling pathways (68), use of PPAR-γ agonists alone on much more common advanced epithelial malignancies has minimal clinical effect (9). Therefore, the combination of PPAR-γ agonists with other drugs, such as EGFR inhibitor (10) or AKT inhibitor (11) has been examined for cancer treatment.

One of the hallmarks of tumor cells is the ability to evade apoptosis (12). Overexpression of antiapoptotic genes is one of mechanisms to escape cancer cell apoptosis. As an important member of the inhibitor of apoptosis gene family, survivin can block the activation of effector caspases in intrinsic and extrinsic pathways of apoptosis. Survivin is absent in normal urothelium, whereas it is present in 64–100% of bladder cancers (13). Moreover, the expression of survivin is associated with high stage and grade as well as with an increase risk of recurrence for patients with bladder cancer (1417). Thus, survivin has been suggested as a suitable target for the development of specific treatment of bladder cancer (15). In the present study, we report that the combination of PPAR-γ activation and survivin inhibition generates a more robust suppression in the cell survival and stem cell properties of bladder cancer cells, providing a basis for future studies testing the strategy for experimental manipulation of bladder cancer.

Materials and methods

Cell culture

The human bladder cancer cell lines, T24 and 5637, obtained from the American Type Culture Collection (Manassas, VA, USA), were cultured in a maintenance medium containing DMEM with high glucose supplemented with 10% fetal bovine serum and penicillin/streptomycin [1% (v/v)] (all from Gibco, Grand Island, NY, USA). The cells were then treated with 15d-PGJ2 (Sigma-Aldrich, St. Louis, MO, USA) for the indicated times (6 days). Cell cultures were maintained at 37°C in a humidified atmosphere with 5% CO2.

siRNA of survivin

Survivin RNAi oligos and negative control high GC oligo were purchased from Sigma-Aldrich. The siRNA sequences are listed in Table I. One day prior to the transfection, T24 and 5637 cells were seeded in 6-well plates without antibiotics. Using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA), the siRNAs (60 nM) were transfected into the cells according to the manufacturer’s instructions.

Table I

List of siRNA sequences.

Table I

List of siRNA sequences.

NamesiRNA sequence
siSurvivin1F GUCUGGACCUCAUGUUGUUdTdT
R AACAACAUGAGGUCCAGACdTdT
siSurvivin2F CCUCUACUGUUUAACAACAdTdT
R UGUUGUUAAACAGUAGAGGdTdT
siSurvivin3F GGUUUAUUCCCUGGUGCCAdTdT
R UGGCACCAGGGAAUAAACCdTdT

[i] F, forward; R, reverse.

Cell viability assay

To evaluate the effect of 15d-PGJ2 on T24 and 5637 cell growth, cell viability was determined by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) (Sigma-Aldrich) assay. In brief, a total of 1,000 5637 or 5,000 T24 cells/well were seeded in 96-well plates (BD Biosciences, San Jose, CA, USA) in a volume of 200 μl. Subsequent to incubation with 0.5 or 2 μg/ml 15d-PGJ2 for the indicated times, 20 μl MTT solution [5 mg/ml in phosphate-buffered saline (PBS)] was added to each well and incubated for an additional 4 h at 37°C. MTT solution was aspirated off, 150 μl dimethyl sulfoxide (DMSO) was added to each well, and the absorbance was measured at 540 nm. Data were recorded on a daily basis and the growth curve was drawn.

Cell cycle analysis

Cells were collected and centrifuged at 300 × g at 4°C for 5 min and resuspended by PBS in tubes. The abovementioned steps were then repeated. The cells were fixed in ice-cold 70% ethanol overnight. After washing with PBS twice, the cells were labelled with propidium iodide (PI) (50 μg/ml; Sigma-Aldrich) and treated with RNase A (100 μg/ml; Amresco, Solon, OH, USA) for 30 min in the dark. The cells were then analyzed using a FACSCalibur flow cytometer (BD Biosciences).

Apoptosis assay

T24 and 5637 cells were collected, centrifuged at 300 × g at 4°C for 5 min, and washed twice with PBS containing 0.5% BSA. The cells were dissociated in 1X binding buffer and the cell concentration was adjusted to 1×106/ml. Cell suspension (100 μl) was added with Annexin V-FITC (BD Biosciences) and 7-AAD (Sigma-Aldrich) according to the manufacturer’s instructions, and incubated for 20 min in the dark. Following the addition of 200 μl 1X binding buffer in the tube, FACS was performed.

Western blot analysis

Cells were lysed in a RIPA lysis buffer (Beyotime, Nantong, China) with Protease Inhibitor Cocktail and PhosSTOP (Roche, Monza, IT, USA). Proteins were detected using indicated antibodies: anti-PPAR-γ, anti-survivin (all from Cell Signaling Technology, Beverly, MA, USA); anti-GAPDH, anti-α-tubulin (both from Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). The ChemiDoc™ XRS system (Bio-Rad Laboratories, Hercules, CA, USA) was used for obtaining images.

Sphere formation assay

To assay sphere formation efficiency, single cells were plated in Ultra Low Attachment plates (Costar, Corning, NY, USA) and cultured in 1:1 DMEM:F12 (Gibco) supplemented with B27 (1:50; Invitrogen), 20 ng/ml epidermal growth factor and 20 ng/ml basic fibroblast growth factor (R&D Systems, Minneapolis, MN, USA). The cells were incubated in a CO2 incubator for 1–2 weeks, and spheres were counted under a stereomicroscope (Olympus, Tokyo, Japan).

Measurement of reactive oxygen species (ROS) accumulation

ROS was monitored by FACS using dihydroethidium (DHE) (Invitrogen). Cells were incubated with 5 μM DHE at 37°C for 30 min, and fluorescence was measured by a FACSCalibur flow cytometer.

Statistical analysis

Data are presented as the means ± SEM. Statistical analyses were conducted using SPSS 13.0 for Windows. Data between two groups were assessed using the Student’s t-test. P <0.05 was considered to indicate statistical significance.

Results

15d-PGJ2 effectively inhibits cell proliferation and stem cell-like properties of bladder cancer cells

Since PPAR-γ agonists are known to inhibit cell growth in various types of cancer cells (1820), we first detected the effect of 15d-PGJ2, a natural PPAR-γ ligand, on the cell viability of bladder cancer cells. T24 and 5637 cells were treated with 15d-PGJ2 at various concentrations for the indicated times (6 days). 15d-PGJ2 efficiently suppressed T24 and 5637 cell growth (Fig. 1A). Similarly, we observed the inhibitory effect of 15d-PGJ2 on bladder cancer cells by measurement of foci formation and cell apoptosis (Fig. 1B and C).

Since PPAR-γ agonists have been observed to regulate differentiation of myxoid/round cell liposarcoma (21) and inhibit tumor-initiating cells in brain and liver cancers (22), we investigated the possibility that 15d-PGJ2 treatment affects the stem cell-like properties of bladder cancer cells. The expression of the stemness-related genes, Oct4 and Nanog, was significantly downregulated in T24 and 5637 cells following the treatment of 15d-PGJ2 (Fig. 2A). Of note, the decreased expression of Oct4 and Nanog genes was observed starting ~1 h after treatment with 15d-PGJ2 in T24 cells, suggesting the key role of 15d-PGJ2 on the repression of the stem-like phenotype of bladder cancer cells. We also performed a speroid formation assay. The results showed that 15d-PGJ2 treatment at low concentrations had no significant effect on the spheroid formation of T24 or 5637 cells (Fig. 2B). Only the treatment with a high dose of 15d-PGJ2 (up to 5 μg/ml) decreased the spheroid number that bladder cancer cells formed (Fig. 2B), suggesting that the treatment of 15d-PGJ2 alone is not sufficient to prevent bladder cancer.

Survivin inhibition accelerates the suppressive effect of 15d-PGJ2 on cell proliferation and the stem cell-like properties of bladder cancer cells

The anti-apoptotic protein survivin has been demonstrated as a promising biomarker for detection and prognosis in bladder cancer. Thus, we hypothesized whether the combination of 15d-PGJ2 and survivin inhibition may more efficiently inhibit cell growth and the stem-like phenotype of bladder cancer cells as compared to the single treatment of 15d-PGJ2. We first evaluated the expression of survivin in 15d-PGJ2-treated bladder cancer cells. The results showed no significant difference in the expression of survivin between the cells treated with or without 15d-PGJ2 (Fig. 3), suggesting that downregulation of survivin may increase the efficiency of 15d-PGJ2 treatment. Survivin expression was depleted with specific siRNAs in the T24 and 5637 cells and the effectiveness of survivin siRNAs was validated by western blotting (Fig. 4A). In the presence of 15d-PGJ2, we found that inhibition of survivin expression by specific siRNAs increased cell apoptosis induced by 15d-PGJ2 (Fig. 4B). Moreover, siRNAs against survivin strengthened the suppressive effect of 15d-PGJ2 on the spheroid formation of T24 cells (Fig. 5A). Notably, the downregulation of survivin by siRNA did not facilitate 15d-PGJ2-mediated inhibition of the stemness-related genes in 5637 cells, even if survivin depletion alone affected the expression of Oct4 and Nanog (Fig. 5B), suggesting that survivin inhibition by siRNAs exacerbated the inhibitory effects of 15d-PGJ2 on bladder cancer cells by directly inducing cell death.

15d-PGJ2 enhances survivin inhibition-induced production of ROS in bladder cancer cells

Since oxidative stress is one of the most important regulatory mechanisms for cell apoptosis and differentiation (2325), we evaluated the generation of ROS in bladder cancer cells treated with 15d-PGJ2 or transfected with survivin-specific siRNAs. Depleting the survivin expression significantly induced the production of ROS in the T24 and 5637 cells, and 15d-PGJ2 further facilitated the generation of ROS (Fig. 5C and D). The upward trend in ROS was consistent with an increase of cell apoptosis induced by 15d-PGJ2 and/or survivin inhibition (Fig. 4B), suggesting that generation of ROS may be responsible for the inhibition of cell proliferation-mediated 15d-PGJ2 and/or survivin in bladder cancer cells.

Discussion

PPAR-γ participates in multiple biological pathways, such as lipid metabolism, energy homeostasis, cell proliferation, death and differentiation (26,27), and various pathogenic processes including inflammation, diabetes, atherosclerosis and cancer (2830). However, despite extensive studies on the PPAR-γ agonists for tumor suppression, the effects of PPAR-γ agonists in tumor-initiating cells (TICs) is still poorly defined. In this study, we showed that 15d-PGJ2, the natural ligand of PPAR-γ, impaired the maintenance and function of TICs in bladder cancer cells. Moreover, the combination of survivin inhibition and 15d-PGJ2 yielded greater inhibition of cultured cell spheroid formation and cell growth of bladder cancer cells.

It is becoming increasingly evident that TICs overexpress multidrug resistance proteins (31,32), which provide a possible explanation for the failure of standard chemotherapy (3336). Our results have demonstrated that 15d-PGJ2 significantly repressed the spheroid formation of bladder cancer cells, decreased the expression of stemness-related genes, indicating that PPAR-γ agonists have a marked inhibitory effect on tumor-initiating cells of human bladder cancer. Survivin is a key biomarker for the detection of bladder cancer metastasis (13,14). When we combined 15d-PGJ2 with survivin depletion, the cell proliferation and spheroid formation were more efficiently suppressed than either alone. These findings raise the possibility that the combination of survivin suppressants and PPAR-γ agonists is likely a new therapy for bladder cancer.

ROS play critical roles in the regulation of cell proliferation, apoptosis, and transformation (24,37). It has recently been established that 15d-PGJ2 negatively regulates cell proliferation by eliciting the production of ROS (3841). More importantly, previous studies have demonstrated that the level of intracellular ROS is associated with TICs (4246). In the present results, we demonstrated that 15d-PGJ2 upregulated the production of ROS, and knockdown of survivin obviously enhanced the generation of ROS stimulated by 15d-PGJ2, suggesting that 15d-PGJ2 and/or survivin inhibition restrained bladder cancer stem-like phenotype and cell proliferation possibly by upregulating ROS production. NADPH oxidases and mitochondria are two major sources of ROS generation (47,48). A recent study has suggested that PPAR-γ agonist may involve mitochondrial function (49). Additionally, it has been shown that PPAR-γ agonists inhibit stem cell-like phenotype and cell proliferation of liver cancer cells via NOX2-mediated oxidative stress (50). Thus, the manner in which the PPAR-γ agonist is involved in the production of ROS induced by 15d-PGJ2 and/or survivin suppression in bladder cancer cells remains to be investigated.

In conclusion, we have shown that cotreatment of 15d-PGJ2 and survivin RNAi synergistically inhibit bladder cancer stem-like phenotype and cell proliferation in vitro. These observations suggest that the combined treatment with survivin inhibitor and PPAR-γ agonists may be of therapeutic importance in the clinical treatment of malignant tumors.

Acknowledgements

This study was funded by the Shanghai Minhang Natural Science Foundation (NO. 2009MHZ109).

References

1 

Ploeg M, Aben KK and Kiemeney LA: The present and future burden of urinary bladder cancer in the world. World J Urol. 27:289–293. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Basile KJ and Aplin AE: Resistance to chemotherapy: short-term drug tolerance and stem cell-like subpopulations. Adv Pharmacol. 65:315–314. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Michalik L, Desvergne B and Wahli W: Peroxisome-proliferator-activated receptors and cancers: complex stories. Nat Rev Cancer. 4:61–70. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Sarraf P, Mueller E, Jones D, et al: Differentiation and reversal of malignant changes in colon cancer through PPARgamma. Nat Med. 4:1046–1052. 1998. View Article : Google Scholar : PubMed/NCBI

5 

Chang TH and Szabo E: Induction of differentiation and apoptosis by ligands of peroxisome proliferator-activated receptor gamma in non-small cell lung cancer. Cancer Res. 60:1129–1138. 2000.PubMed/NCBI

6 

Keshamouni VG, Reddy RC, Arenberg DA, et al: Peroxisome proliferator-activated receptor-γ activation inhibits tumor progression in non-small-cell lung cancer. Oncogene. 23:100–108. 2004.

7 

Chen YC, Shen SC and Tsai SH: Prostaglandin D(2) and J(2) induce apoptosis in human leukemia cells via activation of the caspase 3 cascade and production of reactive oxygen species. Biochim Biophys Acta. 1743:291–304. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Ray DM, Akbiyik F and Phipps RP: The peroxisome proliferator-activated receptor gamma (PPARgamma) ligands 15-deoxy-Delta12,14-prostaglandin J2 and ciglitazone induce human B lymphocyte and B cell lymphoma apoptosis by PPARgamma-independent mechanisms. J Immunol. 177:5068–5076. 2006. View Article : Google Scholar

9 

Girnun GD, Naseri E, Vafai SB, et al: Synergy between PPARgamma ligands and platinum-based drugs in cancer. Cancer Cell. 11:395–406. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Mansure JJ, Nassim R, Chevalier S, et al: A novel mechanism of PPARγ induction via EGFR signalling constitutes rational for combination therapy in bladder cancer. PLoS One. 8:e559972013.

11 

Liu L, Yang Z, Xu Y, et al: Inhibition of oxidative stress-elicited AKT activation facilitates PPARγ agonist-mediated inhibition of stem cell character and tumor growth of liver cancer cells. PLoS One. 8:e730382013.PubMed/NCBI

12 

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

13 

Margulis V, Lotan Y and Shariat SF: Survivin: a promising biomarker for detection and prognosis of bladder cancer. World J Urol. 26:59–65. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Shariat SF, Ashfaq R, Karakiewicz PI, et al: Survivin expression is associated with bladder cancer presence, stage, progression, and mortality. Cancer. 109:1106–1113. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Akhtar M, Gallagher L and Rohan S: Survivin: role in diagnosis, prognosis, and treatment of bladder cancer. Adv Anat Pathol. 13:122–126. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Atlasi Y, Mowla SJ and Ziaee SA: Differential expression of survivin and its splice variants, survivin-DeltaEx3 and survivin-2B, in bladder cancer. Cancer Detect Prev. 32:308–313. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Chen X, Wang T, Yang D, et al: Expression of the IAP protein family acts cooperatively to predict prognosis in human bladder cancer patients. Oncol Lett. 5:1278–1284. 2013.PubMed/NCBI

18 

Lee N, Oh J, Ban J, et al: 4-O-methylhonokiol, a PPARγ agonist, inhibits prostate tumour growth: p21-mediated suppression of NF-κB activity. Br J Pharmacol. 168:1133–1145. 2013.PubMed/NCBI

19 

Smallridge RC, Copland JA, Brose MS, et al: Efatutazone, an oral PPAR-γ agonist, in combination with paclitaxel in anaplastic thyroid cancer: results of a multicenter phase 1 trial. J Clin Endocrinol Metab. 98:2392–2400. 2013.

20 

Ban JO, Oh JH, Son SM, et al: Troglitazone, a PPAR agonist, inhibits human prostate cancer cell growth through inactivation of NFκB via suppression of GSK-3β expression. Cancer Biol Ther. 12:288–296. 2011.PubMed/NCBI

21 

Charytonowicz E, Terry M, Coakley K, et al: PPARγ agonists enhance ET-743-induced adipogenic differentiation in a transgenic mouse model of myxoid round cell liposarcoma. J Clin Invest. 122:886–898. 2012.

22 

Pestereva E, Kanakasabai S and Bright JJ: PPARγ agonists regulate the expression of stemness and differentiation genes in brain tumour stem cells. Br J Cancer. 106:1702–1712. 2012.

23 

Pereira L, Igea A, Canovas B, et al: Inhibition of p38 MAPK sensitizes tumour cells to cisplatin-induced apoptosis mediated by reactive oxygen species and JNK. EMBO Mol Med. 5:1759–1774. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Lambeth JD: NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol. 4:181–189. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Owusu-Ansah E and Banerjee U: Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation. Nature. 461:537–541. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Houseknecht KL, Cole BM and Steele PJ: Peroxisome proliferator-activated receptor gamma (PPARgamma) and its ligands: a review. Domest Anim Endocrinol. 22:1–23. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Kersten S and Wahli W: Peroxisome proliferator activated receptor agonists. EXS. 89:141–151. 2000.PubMed/NCBI

28 

Koshiyama H, Shimono D, Kuwamura N, et al: Rapid communication: inhibitory effect of pioglitazone on carotid arterial wall thickness in type 2 diabetes. J Clin Endocrinol Metab. 86:3452–3456. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Lehmann JM, Moore LB, Smith-Oliver TA, et al: An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma). J Biol Chem. 270:12953–12956. 1995. View Article : Google Scholar : PubMed/NCBI

30 

Su CG, Wen X, Bailey ST, et al: A novel therapy for colitis utilizing PPAR-gamma ligands to inhibit the epithelial inflammatory response. J Clin Invest. 104:383–389. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Kondo T, Setoguchi T and Taga T: Persistence of a small subpopulation of cancer stem-like cells in the C6 glioma cell line. Proc Natl Acad Sci USA. 101:781–786. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Salmaggi A, Boiardi A, Gelati M, et al: Glioblastoma-derived tumorospheres identify a population of tumor stem-like cells with angiogenic potential and enhanced multidrug resistance phenotype. Glia. 54:850–860. 2006. View Article : Google Scholar

33 

Ding W, Mouzaki M, You H, et al: CD133+ liver cancer stem cells from methionine adenosyl transferase 1A-deficient mice demonstrate resistance to transforming growth factor (TGF)-beta-induced apoptosis. Hepatology. 49:1277–1286. 2009.

34 

Hermann PC, Huber SL, Herrler T, et al: Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 1:313–323. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Reya T, Morrison SJ, Clarke MF and Weissman IL: Stem cells, cancer, and cancer stem cells. Nature. 414:105–111. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Kelly PN, Dakic A, Adams JM, et al: Tumor growth need not be driven by rare cancer stem cells. Science. 317:3372007. View Article : Google Scholar : PubMed/NCBI

37 

Adler V, Yin Z, Tew KD and Ronai Z: Role of redox potential and reactive oxygen species in stress signaling. Oncogene. 18:6104–6111. 1999. View Article : Google Scholar : PubMed/NCBI

38 

Kondo M, Oya-Ito T, Kumagai T, et al: Cyclopentenone prostaglandins as potential inducers of intracellular oxidative stress. J Biol Chem. 276:12076–12083. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Kim KY, Ahn JH and Cheon HG: Apoptotic action of peroxisome proliferator-activated receptor-γ activation in human non small-cell lung cancer is mediated via proline oxidase-induced reactive oxygen species formation. Mol Pharmacol. 72:674–685. 2007.

40 

Wang JJ and Mak OT: Induction of apoptosis by 15d-PGJ2 via ROS formation: an alternative pathway without PPARγ activation in non-small cell lung carcinoma A549 cells. Prostaglandins Other Lipid Mediat. 94:104–111. 2011.PubMed/NCBI

41 

Shin SW, Seo CY, Han H, et al: 15d-PGJ2 induces apoptosis by reactive oxygen species-mediated inactivation of Akt in leukemia and colorectal cancer cells and shows in vivo antitumor activity. Clin Cancer Res. 15:5414–5425. 2009.

42 

Ito K, Hirao A, Arai F, et al: Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature. 431:997–1002. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Miyamoto K, Araki KY, Naka K, et al: Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell. 1:101–112. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Tothova Z, Kollipara R, Huntly BJ, et al: FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. 128:325–339. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Yalcin S, Zhang X, Luciano JP, et al: Foxo3 is essential for the regulation of ataxia telangiectasia mutated and oxidative stress-mediated homeostasis of hematopoietic stem cells. J Biol Chem. 283:25692–25705. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Ghaffari S: Oxidative stress in the regulation of normal and neoplastic hematopoiesis. Antioxid Redox Signal. 10:1923–1940. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Alexandre J, Hu Y, Lu W, et al: Novel action of paclitaxel against cancer cells: bystander effect mediated by reactive oxygen species. Cancer Res. 67:3512–3517. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Bedard K and Krause KH: The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 87:245–313. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Zolezzi JM, Silva-Alvarez C, Ordenes D, et al: Peroxisome proliferator-activated receptor (PPAR) γ and PPARα agonists modulate mitochondrial fusion-fission dynamics: relevance to reactive oxygen species (ROS)-related neurodegenerative disorders? PLoS One. 8:e640192013.

Related Articles

Journal Cover

July-2014
Volume 34 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang Y, Tan H, Xu D, Ma A, Zhang L, Sun J, Yang Z, Liu Y and Shi G: The combinatory effects of PPAR-γ agonist and survivin inhibition on the cancer stem-like phenotype and cell proliferation in bladder cancer cells. Int J Mol Med 34: 262-268, 2014
APA
Wang, Y., Tan, H., Xu, D., Ma, A., Zhang, L., Sun, J. ... Shi, G. (2014). The combinatory effects of PPAR-γ agonist and survivin inhibition on the cancer stem-like phenotype and cell proliferation in bladder cancer cells. International Journal of Molecular Medicine, 34, 262-268. https://doi.org/10.3892/ijmm.2014.1774
MLA
Wang, Y., Tan, H., Xu, D., Ma, A., Zhang, L., Sun, J., Yang, Z., Liu, Y., Shi, G."The combinatory effects of PPAR-γ agonist and survivin inhibition on the cancer stem-like phenotype and cell proliferation in bladder cancer cells". International Journal of Molecular Medicine 34.1 (2014): 262-268.
Chicago
Wang, Y., Tan, H., Xu, D., Ma, A., Zhang, L., Sun, J., Yang, Z., Liu, Y., Shi, G."The combinatory effects of PPAR-γ agonist and survivin inhibition on the cancer stem-like phenotype and cell proliferation in bladder cancer cells". International Journal of Molecular Medicine 34, no. 1 (2014): 262-268. https://doi.org/10.3892/ijmm.2014.1774