Open Access

Role of Wnt/β‑catenin, Wnt/c‑Jun N‑terminal kinase and Wnt/Ca2+ pathways in cisplatin‑induced chemoresistance in ovarian cancer

  • Authors:
    • Lu Huang
    • Ye Jin
    • Shujun Feng
    • Yuqing Zou
    • Sainan Xu
    • Shuang Qiu
    • Ling Li
    • Jianhua Zheng
  • View Affiliations

  • Published online on: November 8, 2016     https://doi.org/10.3892/etm.2016.3885
  • Pages: 3851-3858
  • Copyright: © Huang 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

The aim of the present study was to explore the expression of Wnt signaling proteins β-catenin, c-Jun N-terminal kinase (JNK) and Ca2+/calmodulin‑dependent protein kinase II (CaMKII) in ovarian cancer cells, and assess the correlation between this expression and cisplatin‑induced chemoresistance. SKOV3 ovarian carcinoma cells and SKOV3/DDP (cisplatin resistant) cells were treated with cisplatin in the absence or presence of a Wnt signaling activator (CHIR‑99021, glycogen synthase kinase 3β inhibitor) or inhibitor (XAV‑939, tankyrase inhibitor). Following incubation for 48 h, cell viability, proliferation and cytotoxicity were measured using a sensitive colorimetric cell counting kit. Expression levels of β‑catenin, JNK and CaMKII were detected by western blot and immunofluorescence staining. The results of the current study identified that β‑catenin and JNK expression levels were significantly higher (P<0.01 and P<0.05 respectively), while CaMKII expression was lower (P>0.05), in SKOV3/DDP cells compared with SKOV3 cells. Moreover, following treatment with 20 µM cisplatin, reduced expression of β‑catenin and JNK (P<0.05 and P<0.01 respectively), and increased expression of CaMKII (P<0.01), was observed in SKOV3 and SKOV3/DPP cell lines. Furthermore, inhibition of β‑catenin signaling by XAV‑939 effectively reversed cisplatin chemoresistance in SKOV3/DDP cells. Similarly, XAV‑939 downregulated JNK expression (P<0.001), but upregulated CaMKII expression (P<0.001), in SKOV3/DDP cells. In conclusion, abnormal activation of Wnt/β‑catenin and Wnt/JNK signaling pathways in ovarian cancer cells promotes cisplatin resistance, while the Wnt/Ca2+ signaling pathway reduces cisplatin resistance. This indicates that β‑catenin, JNK and CaMKII are potential therapeutic targets in chemoresistant ovarian cancers.

Introduction

Ovarian cancer is the fifth most common cause of death in women worldwide (1). Typical treatment for ovarian cancer involves cytoreductive surgery combined with cisplatin-based chemotherapy (2). Unfortunately, the initial response rate of this treatment is not sustainable and in 70% of women disease reoccurs within months as a result of chemoresistance (3,4). It has been suggested that the high mortality rate in ovarian cancer can be partly attributed to this high frequency of chemoresistance (5).

Cisplatin is a platinum compound that was found to arrest binary fission in Escherichia coli in the 1960s, and it is now a crucial chemotherapeutic drug in the treatment of numerous cancers, including ovarian, as a single agent and in combination with other anticancer drugs (6,7). The issue of resistance to cisplatin remains a major obstacle in the successful treatment of ovarian cancer (8). Previous studies have indicated that inhibition of intrinsic cell death signaling pathways, activation of cell survival signaling pathways, and dysregulation of oncogenes, tumor suppressor genes and microRNAs contributes to cisplatin chemoresistance in ovarian cancer cells (9,10). However, the underlying mechanisms by which cisplatin chemoresistance occurs in ovarian cancer cells remain unclear.

Previous studies have identified that abnormal Wnt signaling serves a role in the development of breast (11), gastric (12), lung (13), prostate (14) and endometrial (15) cancers. In ovarian cancer, persistent activation of Wnt signaling was observed to increase cell survival, and several studies have concluded that aberrant regulation of Wnt signaling induces tumor cell chemoresistance (1618).

The Wnt signaling signaling pathway serves a critical role in embryogenesis and oncogenesis (19). Wnt ligands are a family of secreted proteins comprising of 19 members (10). Wnt ligands bind to a Frizzled (Fzd) family receptor and low-density lipoprotein receptor-related protein (LRP)-5/6 to initiate signaling (10). Wnt signaling is divided into canonical Wnt signaling pathway (Wnt/β-catenin) and non-canonical Wnt signaling pathways (19).

In the canonical Wnt signaling pathway, illustrated in Fig. 1A, Wnt ligands engage Fzd and LRP-5/6, which inhibits glycogen synthase kinase 3β (GSK-3β), leading to stabilization and increasing expression levels of β-catenin in the cytoplasm. Stable β-catenin translocates into the nucleus where it binds to the transcription factor T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) to control the transcription of Wnt target genes (20).

Dysregulation of the Wnt/β-catenin signaling pathway has been identified in numerous cancers, including ovarian (21). β-catenin is the primary component of this signaling pathway, and mutations in the gene encoding β-catenin (CTNNB1), leading to alteration of the Wnt/β-catenin signaling pathway, have been found in the endometrioid subtype of ovarian cancer (22,23). Furthermore, aberrant expression of β-catenin and Wnt-5A has been observed in ovarian cancer (22,24). In addition, aberrant accumulation of β-catenin is associated with increasing ovarian cancer grade and poor survival (25,26). However, the use of β-catenin as a prognostic marker for ovarian cancer is disputed. To the best of our knowledge, the impact of β-catenin expression levels on chemoresistance has not been evaluated in ovarian cancer.

The non-canonical Wnt signaling pathways involve signaling that uses downstream effectors, without mediation by β-catenin-TCF/LEF. In contrast to canonical Wnt signaling, these signaling pathways may have transcriptional and non-transcriptional effects (27). In the non-canonical Wnt/Ca2+ signaling pathway, Wnt ligands binding to Fzd receptors initiates the activation of phospholipase C via G protein-couple receptor signaling, causing an increase in intracellular Ca2+, resulting in activation of Ca2+/calmodulin-dependent kinase II (CaMKII) and protein kinase C. Previous studies have identified that deregulation of the Wnt/Ca2+ signaling pathway mediates cytoskeleton rearrangements, cellular proliferation, cellular motility and epithelial-mesenchymal transition in cancer development and progression (Fig. 1B) (27).

In the non-canonical Wnt/JNK signaling pathway, Wnt-Fzd binding activates a number of small guanosine-5′-triphosphate (GTP) -binding proteins, including c-Jun N-terminal kinase (JNK), which affect a wide range of cellular processes, including cytoskeleton rearrangement, cell polarity, cell migration and gene expression (28). Previous studies have identified that aberrant Wnt/JNK signaling may initiate and stimulate the development of malignant phenotypes through effects on cell proliferation, survival, polarity, invasion and metastasis (Fig. 1C) (29,30).

Aberrant Wnt/β-catenin, Wnt/JNK and Wnt/Ca2+ signaling has been associated with increasing ovarian cancer grade and poor survival (21,27,29). However, to the best of our knowledge, the relationship between these signaling pathways and ovarian cancer cisplatin chemoresistance has not yet been investigated (23,29).

The present study aims to determine the expression of β-catenin, JNK and CaMKII in a cisplatin-sensitive ovarian cancer cell line and a cisplatin-resistant variant, and then assess the correlation between expression of these proteins and cisplatin chemoresistance.

Materials and methods

Chemicals and reagents

Cisplatin was purchased from Sigma-Aldrich (Merck Millipore, Darmstadt, Germany). Cell counting kit-8 (CCK-8; C0037) and 4′,6-diamidino-2-phenylindole (DAPI; C1005) were purchased from the Beyotime Institute of Biotechnology (Haimen, China) and stored at −20°C. Antibodies including non-phospho (active) β-catenin (Ser33/37/Thr41) (D13A1) rabbit mAb (#8814), SAPK/JNK rabbit mAb (#9252) and CaMKII (pan) (D11A10) Rabbit mAb (#4436) were purchased from Cell Signaling Technology, Inc. (Beverly, MA, USA), and the β-actin mouse mAb (JT9001S) was purchased from Anbo Biotechnology Co., Ltd, (San Francisco, CA, USA). The Wnt signaling inhibitor (XAV-939) and activator (CHIR-99021) were purchased from the Selleck Chemicals (Shanghai, China), and completely dissolved in DMSO at a stock concentration (1 mM). Fluorescein isothiocyanate (FITC)-Conjugated AffiniPure Goat Anti-Mouse IgG (H+L) (ZF-0312) and FITC-Conjugated AffiniPure Goat Anti-Rabbit IgG (H+L) (ZF-0315) were from OriGene Technologies, Inc. (Beijing, China).

Cell culture

The human ovarian cancer adenocarcinoma cell line SKOV3 and its cisplatin-resistant variant, SKOV3/DDP, were purchased from The Chinese Academy of Sciences (Shanghai, China). Cells were cultured in Roswell Park Memorial Institute (RPMI)-1640 media, supplemented with 100 U/ml penicillin/streptomycin and 10% fetal bovine serum (FBS) at 37°C and 5% CO2. In addition, the SKOV3/DDP cell media included 2 µM cisplatin to maintain chemoresistance. All of the described reagents were purchased from GE Healthcare Life Sciences (HyClone; Logan, UT, USA).

Cell viability and cytotoxicity assay

Cell viability was detected using the CCK-8 kit. Briefly, cells were seeded at 5×103 cells/well into 96-well plates. Following 48 h incubation as same as previously described conditions, cells were treated with cisplatin, CHIR-99021, XAV-939, cisplatin + CHIR-99021 or cisplatin + XAV-939 for indicated concentrations. There were six replicate wells for each concentration, the control group contained untreated cells and in the blank group (no cells or drugs) only added 10% FBS and RMPI-1640. Cells were incubated for 24, 48 or 72 h prior to the addition of 10 µl CCK-8 reagent and 90 µl RMPI-1640 to each well and further culture for 1 h. The absorbance of each well at 490 nm (A490) was then analyzed using a microplate reader (Model 680; Bio-Rad Laboratories, Inc., Hercules, CA, USA). The effect of the drugs on the cell survival was calculated using the following formula: Survival rate (%) = [(A490 of treated group - A490 of blank group) / (A490 of control group - A490 of blank group)] × 100. The inhibition rate (%) was calculated as: (100%-survival rate %). For the concentration of cisplatin necessary to result in 50% inhibition (IC50), the data was calculated using the weighted linear regression method with SPSS 22.0 software (IBM SPSS, Armonk, NY, USA). Each experiment was repeated >3 times.

Western blotting

Cells were treated with cisplatin (20 µM), CHIR-99021 (3 µM), XAV-939 (1 µM), cisplatin + CHIR-99021 (20 and 3 µM, respectively), cisplatin + XAV-939 (20 and 1 µM, respectively) or RPMI-1640 alone for 48 h, as previously described. Then, total protein was extracted by treatment with RIPA lysis buffer (P0013B; Beyotime Institute of Biotechnology) for 45 min on ice, centrifugation (10,000 × g) for 25 min at 4°C and collection of the supernatants, which were stored at −20°C until required. Total protein concentration was determined using the BCA Protein Assay Kit (P0010S; Beyotime Institute of Biotechnology). Equal quantities of protein preparations (50 µg) were separated by 12% SDS-PAGE and transferred onto polyvinylidene fluoride membranes. The membranes were then blocked with 5% non-fat milk at room temperature for between 1 and 2 h and incubated overnight with primary antibodies against the following proteins: β-catenin (1:1,000), JNK (1:2,000), CaMKII (1:1,000) and β-actin (1:5,000). Next, membranes were incubated with the appropriate fluorescently-labeled secondary antibodies (1:10,000). Between each step, protein bands were washed with 1X PBST. Protein bands were detected using an Odyssey Infrared Imaging System (Gene Company, Ltd., Hong Kong, China). Quantitative analysis was performed using ImageJ analysis software (National Institutes of Health, USA) with β-actin serving as a control.

Immunofluorescence staining

Cells were cultured at 2×104 cells/well into 24-well plates for 48 h and treated with cisplatin, CHIR-99021, XAV-939, cisplatin + CHIR-99021, cisplatin + XAV-939 or RPMI-1640 alone for 48 h, as previously described. Cells were then fixed with 4% paraformaldehyde, permeabilized with 0.3% Triton X-100 and blocked with 5% horse serum (Vector Laboratories, Inc., Burlingame, CA, USA) in PBS-Tween for 30 min. Preparations were incubated with primary antibodies [β-catenin (1:200), JNK (1:200) and CaMKII (1:400)] as previously described at 4°C overnight and then with the appropriate fluorescently-labeled secondary antibodies (previously described; 1:1,000) for 60 min at room temperature. In addition, nuclei were stained with DAPI. Between each step, protein bands were washed with 1X PBST. Preparations were visualized and images acquired using the EVOS XL Cell Imaging System (Thermo Fisher Scientific, Inc., Waltham, MA, USA).

Statistical analysis

Data are presented as means ± the standard deviation. Data was analyzed using SPSS software (version 22.0). Differences between groups were statistically analyzed using the Student's t-test or a paired t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Cisplatin sensitivity in SKOV3 and SKOV3/DDP cell lines

To determine the sensitivity of SKOV3 and SKOV3/DDP cells to cisplatin, cells were exposed to a gradient of cisplatin concentrations (0–80 µM) for 24, 48 or 72 h and cell viability was measured using the CCK-8 kit. The inhibition rate of SKOV3 and SKOV3/DDP cells with cisplatin was found to increase in a time-dependent and dose-dependent manner (Fig. 2A-C). There was a significant difference between the concentrations of cisplatin required for IC50 after 48 h in SKOV3 and SKOV3/DDP cells (6.086±0.38 vs. 26.135±1.825 µM, respectively; P<0.05; Fig. 2D), highlighting that SKOV3/DDP cells were significantly cisplatin-resistant compared with SKOV3 cells. From the analysis of these results, a dose of 20 µM cisplatin and an exposure time of 48 h were used in subsequent studies.

Expression of β-catenin, JNK and CaMKII in SKOV3 and SKOV3/DDP cell lines

To determine if cisplatin chemoresistance seen in ovarian cancer cells was associated with deregulation of Wnt signaling pathways, the present study evaluated the protein expression levels of β-catenin, JNK and CaMKII in SKOV3 and SKOV3/DDP cell lines. Quantification of western blotting results (Fig. 3A) identified that expression levels of β-catenin and JNK were significantly higher in SKOV3/DDP cells compared with SKOV3 cells (*P<0.01, &P<0.05; Fig. 3B). However, CaMKII protein expression levels were lower in SKOV3/DPP cells compared with SKOV3 cells. Following incubation with 20 µM cisplatin for 48 h, β-catenin expression was reduced in SKOV3 and SKOV3/DDP cells (*P<0.01, *P<0.001; Fig. 3B). Similarly, JNK protein expression levels were significantly decreased in SKOV3 and SKOV3/DDP cells following treatment with 20 µM cisplatin (&P<0.05, #P<0.001; Fig. 3B). In contrast, cisplatin treatment significantly increased CaMKII protein expression levels in SKOV3 and SKOV3/DDP cells (#P<0.001, *P<0.01; Fig. 3B). The difference in CaMKII expression between SKOV3 and SKOV3/DDP cells was significant following cisplatin treatment (&P<0.05; Fig. 3B). Similar results were observed in immunofluorescence stains (Fig. 3C-E). These results suggest that abnormal activation of Wnt/β-catenin and Wnt/JNK signaling pathways, and the abnormal inactivation of the Wnt/Ca2+ signaling pathway, may be associated with cisplatin chemoresistance in SKOV3/DPP cells.

Association between Wnt/β-catenin, Wnt/JNK and Wnt/Ca2+ signaling pathways and ovarian cancer cisplatin-resistance

To confirm the association observed between the deregulation of Wnt signaling pathways and cisplatin-resistance in ovarian cancer cells, CHIR-99021 (glycogen synthase kinase 3β inhibitor, GSK3β) or XAV-939 (tankyrase inhibitor) were used in addition to cisplatin to specifically activate or inhibit the Wnt/β-catenin signaling pathway, respectively. The effect of these drugs on SKOV3/DPP cell proliferation was then examined using the CCK-8 kit. Compared with the control group without any drugs, the majority of doses of CHIR-99021 or XAV-939 could not induce significant cell death (Fig. 4A and B). However, 3 µM CHIR-99021 combined with cisplatin significantly suppressed the cytotoxicity of cisplatin (P<0.05; Fig. 4A) and 1 µM XAV-939 combined with cisplatin significantly increased the cytotoxicity of cisplatin (P<0.05; Fig. 4B).

Next, SKOV3/DDP cells were treated with or without cisplatin in the presence or absence of 3 µM CHIR-99021 or 1 µM XAV-939, and the expression levels of β-catenin, JNK and CaMKII were examined by western blotting and immunofluorescence staining. Compared with the untreated group, the level of β-catenin was increased by CHIR-99021 treatment (Fig. 4C and D). Futhermore, CHIR-99021 significantly reversed the loss of β-catenin seen in cisplatin-treated cells (&P<0.05; Fig. 4D), reduced cisplatin-induced loss of JNK (&P<0.05) and significantly reduced cisplatin-induced upregulation of CaMKII (#P<0.001; Fig. 4D). In contrast, XAV-939 significantly decreased expression levels of β-catenin and JNK (#P<0.001), and significantly increased expression levels of CaMKII in cells (*P<0.01), with and without co-treatment of cisplatin (Fig. 4E and F). Interestingly, the expression levels of β-catenin and JNK were positively correlated, and the level of CaMKII was negatively correlated with β-catenin and JNK expression levels (Fig. 4E and F). Similar effects of CHIR-99021 and XAV-939 were also observed in immunofluorescence stains (Fig. 5). These results indicate that the Wnt/β-catenin signaling pathway activation antagonizes cisplatin-induced cell death, while the Wnt/β-catenin signaling pathway inhibition enhances the cytotoxic effect of cisplatin on ovarian cancer cells.

Discussion

Ovarian cancer is the leading cause of mortality among gynecological cancers (1). In addition to cytoreductive surgery, cisplatin-based chemotherapy is one of the most important therapeutic strategies for ovarian cancer (2). Previous studies have shown that Wnt signaling pathway deregulation correlates with ovarian cancer cell proliferation, survival, invasion and metastasis (25,26,28,30,31). Furthermore, cisplatin chemoresistance has been an obstacle in the successful treatment of ovarian cancer (8,32,33). Several studies have concluded that aberrant regulation of Wnt signaling induces tumor cell chemoresistance (1618). However, the molecular mechanisms by which Wnt signaling leads to cisplatin chemoresistance remain poorly understood.

In the present study, a specific Wnt/β-catenin signaling pathway activator (CHIR-99021) and inhibitor (XAV-939) were used to explore the association between the deregulation of Wnt signaling and cisplatin-resistance in ovarian cancer cells. Firstly, the human ovarian cancer adenocarcinoma cell line SKOV3 and its cisplatin-resistant variant, SKOV3/DDP, were selected as cell models. The IC50 value of cisplatin in SKOV3/DDP cells was approximately four-fold that of SKOV3 cells, showing that SKOV3/DDP cells were more cisplatin-resistant. Thus, these cell lines are suitable models for the exploring cisplatin chemoresistance.

Previous studies of the endometrioid subtype of ovarian cancer have found mutations of CTNNB1 and abnormal expression of β-catenin (22,23). The present study demonstrated that β-catenin and JNK expression levels were increased in cisplatin-resistant compared with cisplatin-sensitive ovarian cancer cells. In contrast, CaMKII expression levels were lower in cisplatin-resistant ovarian cancer cells.

The correlation of the Wnt/β-catenin and non-canonical Wnt signaling pathways in cisplatin chemoresistance is controversial. Some investigators believe that both canoncial and noncanoncial Wnt signaling pathways may promote each other in cisplatin chemoresistance (34). However, other research suggests that the Wnt/β-catenin signaling pathway is crucial to cisplatin chemoresistance and that the non-canonical Wnt signaling pathways obstruct this action (3537). In the current study, the Wnt/β-catenin signaling pathway agonist CHIR-99021, which specifically inhibits GSK3β, was used to investigate the correlation between the Wnt/β-catenin and non-canonical Wnt signaling pathways. The SKOV3/DPP cell inhibition rate with CHIR-99021 in combination with cisplatin was lower compared with that of cisplatin alone. The level of β-catenin was increased as a result of CHIR-99021 treatment, due to Wnt/β-catenin signaling pathway activation. In addition, CHIR-99021 in combination with cisplatin enhanced expression of JNK and significantly decreased the expression of CaMKII compared with cisplatin alone. Similarly, Zhao et al (21) found an association between high expression levels of β-catenin and cisplatin chemoresistance in A2780/DDP cells. This suggests that interference with β-catenin expression could partly reverse cisplatin chemoresistance in ovarian cancer cells.

SKOV3/DDP cells co-treated with XAV-939, a selective Wnt/β-catenin signaling pathway inhibitor, and cisplatin produced a greater inhibition of proliferation than when applied alone. Expression of β-catenin and JNK was significantly reduced, and expression of CaMKII was significantly higher, in SKOV3/DPP cells following co-treatment with XAV-939 and cisplatin compared with untreated cells or cells treated with cisplatin alone. Furthermore, the expression levels of β-catenin and JNK were positively correlated, while the level of CaMKII was negatively correlated with of β-catenin and JNK expression levels.

In conclusion, the results of the present study identify that, in SKOV3/DDP cells, the Wnt/β-catenin and Wnt/JNK signaling pathways are positively correlated with cisplatin chemoresistance, while the Wnt/Ca2+ signaling pathway is negatively correlated with cisplatin chemoresistance. This suggests that inhibiting the Wnt/β-catenin and Wnt/JNK signaling pathways, and activating the Wnt/Ca2+ signaling pathway, could reverse cisplatin-resistance in ovarian cancer cells. Developing specific inhibitors and activators for these signaling pathways may provide a treatment for cisplatin-resistant ovarian cancer, and the key players of canonical (β-catenin) and non-canonical (JNK and CaMKII) Wnt signaling are potential targets for drug development.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Bristow R, Chang J, Ziogas A, et al: NCCN treatment guidelines for ovarian cancer: A population-based vali-dation study of structural and process quality measures. Gynecologic Oncology. 130:e182013. View Article : Google Scholar

3 

Seiya S and Hiroaki I: Ovarian cancer and drug resistance. Current Obstetrics and Gynecology Reports. 4:18–25. 2015. View Article : Google Scholar

4 

Lloyd KL, Cree IA and Savage RS: Prediction of resistance to chemotherapy in ovarian cancer: A systematic review. BMC Cancer. 15:1172015. View Article : Google Scholar : PubMed/NCBI

5 

Leamon PC, Lovejoy CD and Nguyen B: Patient selection and targeted treatment in the management of platinum-resistant ovarian cancer. Pharmgenomics Pers Med. 6:113–125. 2013.PubMed/NCBI

6 

Go RS and Adjei AA: Review of the comparative pharmacology and clinical activity of cisplatin and carboplatin. J Clin Oncol. 17:409–422. 1999.PubMed/NCBI

7 

Kelland L: The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer. 7:573–584. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Claudia M, Jonathan AL and Pierluigi BP: An overview of early investigational therapies for chemoresistant ovarian cancer. Expert Opin Investig Drugs. 24:1163–1183. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M and Kroemer G: Molecular mechanisms of cisplatin resistance. Oncogene. 31:1869–1883. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Sorrentino A, Liu CG, Addario A, Peschle C, Scambia G and Ferlini C: Role of microRNAs in drug-resistant ovarian cancer cells. Gynecol Oncol. 111:478–486. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Bayerlová M, Klemm F, Kramer F, Pukrop T, Beißbarth T and Bleckmann A: Newly constructed network models of different WNT signaling cascades applied to breast cancer expression data. PLoS One. 10:e01440142015. View Article : Google Scholar : PubMed/NCBI

12 

Shin H, Kim JH, Lee YS and Lee YC: Change in gene expression profiles of secreted frizzled-related proteins (SFRPs) by sodium butyrate in gastric cancers: Induction of promoter demethylation and histone modification causing inhibition of Wnt signaling. Int J Oncol. 40:1533–1542. 2012.PubMed/NCBI

13 

Xi Y and Chen Y: Wnt signaling pathway: Implications for therapy in lung cancer and bone metastasis. Cancer Lett. 353:8–16. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Sidaway P: Prostate cancer: Wnt signaling induces resistance. Nat Rev Urol. 12:5972015. View Article : Google Scholar : PubMed/NCBI

15 

Dellinger T, Warden C, Han E, et al: Wnt pathway gene expression and association with clinicopathologic characteristics in endometrial cancer-An analysis of The Cancer Genome Atlas (TCGA). Gynecologic Oncology. 130:e892013. View Article : Google Scholar

16 

Rosanò L, Cianfrocca R, Tocci P, Spinella F, Di Castro V, Caprara V, Semprucci E, Ferrandina G, Natali PG and Bagnato A: Endothelin A receptor/β-arrestin signaling to the Wnt pathway renders ovarian cancer cells resistant to chemotherapy. Cancer Res. 74:7453–7464. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Su HY, Lai HC, Lin YW, Liu CY, Chen CK, Chou YC, Lin SP, Lin WC, Lee HY and Yu MH: Epigenetic silencing of SFRP5 is related to malignant phenotype and chemoresistance of ovarian cancer through Wnt signaling pathway. Int J Cancer. 127:555–567. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Laura R, Roberta C, Piera T, et al: Endothelin A receptor/β-arrestin signaling to the Wnt pathway renders ovarian cancer cells resistant to chemotherapy. Cancer Research. 74:7463–7464. 2014.

19 

Dawson K, Aflaki M and Nattel S: Role of the Wnt-Frizzled system in cardiac pathophysiology: A rapidly developing, poorly understood area with enormous potential. J Physiol. 591:1409–1432. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Chiurillo MA: Role of the Wnt/β-catenin pathway in gastric cancer: An in-depth literature review. World J Exp Med. 5:84–102. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Zhao H, Wei W, Sun Y, Gao J, Wang Q and Zheng J: Interference with the expression of β-catenin reverses cisplatin resistance in A2780/DDP cells and inhibits the progression of ovarian cancer in mouse model. DNA Cell Biol. 34:55–62. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Barghout SH, Zepeda N, Xu Z, Steed H, Lee CH and Fu Y: Elevated β-catenin activity contributes to carboplatin resistance in A2780cp ovarian cancer cells. Biochem Biophys Res Commun. 468:173–178. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Arend RC, Londoño-Joshi AI, Straughn JM Jr and Buchsbaum DJ: The Wnt/β-catenin pathway in ovarian cancer: A review. Gynecol Oncol. 131:772–779. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Matsumoto T, Yamazaki M, Takahashi H, Kajita S, Suzuki E, Tsuruta T and Saegusa M: Distinct β-Catenin and PIK3CA mutation profiles in endometriosis-associated ovarian endometrioid and clear cell carcinomas. Am J Clin Pathol. 144:452–463. 2015. View Article : Google Scholar : PubMed/NCBI

25 

McConechy MK, Ding J, Senz J, Yang W, Melnyk N, Tone AA, Prentice LM, Wiegand KC, McAlpine JN, Shah SP, et al: Ovarian and endometrial endometrioid carcinomas have distinct CTNNB1 and PTEN mutation profiles. Mod Pathol. 27:128–134. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Forda CE, Punnia-Moorthya G, Henrya CE, Llamosas E, Nixdorf S, Olivier J, Caduff R, Ward RL and Heinzelmann-Schwarz V: The non-canonical Wnt ligand, Wnt5a, is upregulated and associated with epithelial to mesenchymal transition in epithelial ovarian cancer. Gynecol Oncol. 134:338–345. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Niehrs C: The complex world of WNT receptor signaling. Nat Rev Mol Cell Biol. 13:767–779. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Cizelsky W, Tata A, Kühl M and Kühl SJ: The Wnt/JNK signaling target gene alcam is required for embryonic kidney development. Development. 141:2064–2074. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Tong XG, Barbourb M, Houc K, Gao C, Cao S, Zheng J, Zhao Y, Mu R and Jiang HR: Interleukin-33 predicts poor prognosis and promotes ovarian cancer cell growth and metastasis through regulating ERK and JNK signaling pathways. Mol Oncol. 10:113–125. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Ye XL, Zhao YR, Weng GB, Chen YC, Wei XN, Shao JP and Ji H: IL-33-induced JNK pathway activation confers gastric cancer chemotherapy resistance. Oncol Rep. 33:2746–2752. 2015.PubMed/NCBI

31 

De A: Wnt/Ca2+ signaling pathway: A brief overview. Acta Biochim Biophys Sin (Shanghai). 43:745–756. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Ali AY, Farrand L, Kim JY, Byun S, Suh JY, Lee HJ and Tsang BK: Molecular determinants of ovarian cancer chemoresistance: New insights into an old conundrum. Ann N Y Acad Sci. 1271:58–67. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Poisson LM, Munkarah A, Madi H, Datta I, Hensley-Alford S, Tebbe C, Buekers T, Giri S and Rattan R: A metabolomic approach to identifying platinum resistance in ovarian cancer. J Ovarian Res. 8:132015. View Article : Google Scholar : PubMed/NCBI

34 

Luna-Ulloa LB, Hernández-Maqueda JG, Castañeda-Patlán MC and Robles-Flores M: Protein kinase C in Wnt signaling: Implications in cancer initiation and progression. IUBMB Life. 63:915–921. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Kagermeier-Schenk B, Wehner D, Özhan-Kizil G, Yamamoto H, Li J, Kirchner K, Hoffmann C, Stern P, Kikuchi A, Schambony A and Weidinger G: Waif1/5T4 inhibits Wnt/β-catenin signaling and activates noncanonical Wnt pathways by modifying LRP6 subcellular localization. Dev Cell. 21:1129–1143. 2011. View Article : Google Scholar : PubMed/NCBI

36 

King TD, Zhang W, Suto MJ and Li Y: Frizzled7 as an emerging target for cancer therapy. Cell Signal. 24:846–851. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Ishitani T, Kishida S, Hyodo-Miura J, Ueno N, Yasuda J, Waterman M, Shibuya H, Moon RT, Ninomiya-Tsuji J and Matsumoto K: The TAK1-NLK mitogen-activated protein kinase cascade functions in the Wnt-5a/Ca(2+) pathway to antagonize Wnt/beta-catenin signaling. Mol Cell Biol. 23:131–139. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 12 Issue 6

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
Huang L, Jin Y, Feng S, Zou Y, Xu S, Qiu S, Li L and Zheng J: Role of Wnt/β‑catenin, Wnt/c‑Jun N‑terminal kinase and Wnt/Ca2+ pathways in cisplatin‑induced chemoresistance in ovarian cancer. Exp Ther Med 12: 3851-3858, 2016
APA
Huang, L., Jin, Y., Feng, S., Zou, Y., Xu, S., Qiu, S. ... Zheng, J. (2016). Role of Wnt/β‑catenin, Wnt/c‑Jun N‑terminal kinase and Wnt/Ca2+ pathways in cisplatin‑induced chemoresistance in ovarian cancer. Experimental and Therapeutic Medicine, 12, 3851-3858. https://doi.org/10.3892/etm.2016.3885
MLA
Huang, L., Jin, Y., Feng, S., Zou, Y., Xu, S., Qiu, S., Li, L., Zheng, J."Role of Wnt/β‑catenin, Wnt/c‑Jun N‑terminal kinase and Wnt/Ca2+ pathways in cisplatin‑induced chemoresistance in ovarian cancer". Experimental and Therapeutic Medicine 12.6 (2016): 3851-3858.
Chicago
Huang, L., Jin, Y., Feng, S., Zou, Y., Xu, S., Qiu, S., Li, L., Zheng, J."Role of Wnt/β‑catenin, Wnt/c‑Jun N‑terminal kinase and Wnt/Ca2+ pathways in cisplatin‑induced chemoresistance in ovarian cancer". Experimental and Therapeutic Medicine 12, no. 6 (2016): 3851-3858. https://doi.org/10.3892/etm.2016.3885