SPARCL1 suppresses the proliferation and migration of human ovarian cancer cells via the MEK/ERK signaling

  • Authors:
    • Yan Ma
    • Yuan Xu
    • Li Li
  • View Affiliations

  • Published online on: August 7, 2018     https://doi.org/10.3892/etm.2018.6575
  • Pages: 3195-3201
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ovarian cancer is the most lethal gynecological malignancy worldwide and is one of the five leading causes of cancer‑associated mortality in women. There is an urgent requirement to obtain a greater understanding of the molecular mechanism underlying ovarian cancer progression in order to identify novel drug targets and biomarkers. Secreted protein acidic and rich in cysteine‑like protein 1 (SPARCL1) has been suggested as a candidate tumor suppressor in various types of human cancers. However, the potential role of SPARCL1 for ovarian cancer has not yet been clearly established. In the present study, lower protein expression levels of SPARCL1 were detected in ovarian cancer tissues when compared with adjacent normal tissues. Overexpression of SPARCL1 significantly suppressed the proliferation and migration of cells from the ovarian cancer cell line SKOV‑3, whereas knockdown of SPARCL1 significantly increased cell growth and migration. Furthermore, the results revealed that SPARCL1 overexpression significantly suppressed the activation of the mitogen‑activated protein kinase kinase (MEK)/extracellular signal‑related kinase (ERK) signaling pathway. Collectively, these results indicated that SPARCL1 may suppress the proliferation and migration of ovarian cancer cells by downregulating signaling via the MEK/ERK pathway.

Introduction

Ovarian cancer is the most lethal gynecological malignancy and the fifth leading cause of cancer-associated deathin women (1,2). Ovarian cancers are responsible for more than half of the female genital tract cancers caused deaths, with more than 2 million new cases being diagnosed every year (1,3,4). Despite significant advances in surgery and chemotherapy, the long-term survival rate of patients with ovarian cancers is still very low (3). Thus, there is clearly an urgent need to unravel the molecular mechanisms of ovarian cancer progression for an improved treatment outcome.

Secreted protein acidic and rich in cysteine-like protein 1 (SPARCL1) is a member of the SPARC family, which is associated with various biological behaviors including osteoblast differentiation and development of cancers. Previous studies have suggested SPARCL1 as a potential tumor suppressor (57). Low SPARCL1 expression is associated with the development and progression of cervical cancer (8). Zhao et al (5) detected that SPARCL1 was significantly down-regulated in human osteosarcoma cell lines and clinical tissue samples. Interestingly, a recent study revealed that SPARCL1 was strongly down-regulated by 6.4-fold in 594 ovarian cancer cases compared with eight normal ovaries and was suspected to contribute to tumor invasiveness (9). However, the effect of SPARCL1 on ovarian cancer and its underlying mechanisms remain elusive.

The present study aimed to investigate the role of SPARCL1 in ovarian cancer. In this study we are the first to evaluate the expression levels of SPARCL1 in human ovarian cancer tissues and the specific effects of SPARCL1 on ovarian cancer cell proliferation and migration, and to explore possible mechanisms of action.

Materials and methods

Clinical tissues

The present study was approved by the Medical Ethics Committee of the Third Affiliated Hospital, Xinjiang Medical University (Urumqi, China). Signed written informed consent was obtained from all subjects. Twenty ovarian cancer tissues and paired adjacent normal tissues were collected during surgery from ovarian cancer patients without prior chemotherapy or radiotherapy. Disease status had been confirmed in all specimens by pathological diagnosis.

Immunohistochemistry (IHC)

IHC staining was performed using the standard immunoperoxidase staining procedure as previously reported (1012), to detect SPARCL1 expression in paraffin-embedded specimens. The slides of 3 µm thickness were incubated overnight with polyclonal rabbit anti-SPARCL1 antibody (1:100; Abcam, Cambridge, UK). After a secondary antibody was applied for 30 min at room temperature, the slides were stained with diaminobenzidine (DAB) and hematoxylin. The samples were blinded during the independent assessment of immunostaining intensity by three senior pathologists. Each section was photographed and semi-quantitative analyzed using computerized image analysis (Image J; National Institutes of Health, Bethesda, MD, USA).

Cell culture

The human ovarian cancer cell line SKOV-3 was purchased commercially from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). The cells were grown in RPMI-1640 medium (Sigma, Shanghai, China) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), 50 µg/ml streptomycin and 50 IU/ml penicillin, according to the instructions of the American Type Culture Collection (ATCC, Manassas, VA, USA). Cells were cultured at 37°C in a humidified atmosphere of 5% CO2 and were regenerated every 3 days when they reached 70–90% confluence.

Lentiviral transfection

A lentiviral short hairpin RNA (shRNA) construct targeting SPARCL1 (with targeting sequences to SPARCL1 mRNA as follows: 5′-CCCGACAAATGCAAGATTATT-3′) was obtained from Jikai Corporation (Jikai, Shanghai, China). The oligonucleotides were then phosphorylated, annealed, and cloned into the pLKO.1 (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) following the manufacturer's instructions. The overexpression particles of SPARCL1 were purchased from GenePharma (Shanghai, China). Transfection of SPARCL1 was performed following the manufacturer's instructions.

Cell proliferation

The cell proliferation was assessed using a MTT assay kit (Beyotime Institute of Biotechnology, Haimen, China). To evaluate the effects of SPARCL1 on human ovarian cells, SKOV-3 cells (5,000 cells/well in 100 µl medium) were seeded into 96-well plates on days 1, 2, 3, and 4 post-transfection. After 24 h, the medium was removed, and replaced with 100 µl RPMI-1640 medium without FBS supplemented with 20 µl MTT (5 mg/ml). After incubation for 4 h at 37°C, the absorbance at 450 nm was determined using a microplate reader (iMark; Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Scratch wound healing assay

SKOV-3 cells (1×106 cells/well) were seeded into 6-well plates and cultured to a 100% confluent monolayer. In each well a wound was induced by scratching a straight line into the cell layer with a 200 µl pipette tip. After 24 h of incubation with RPMI 1640 medium without FBS, images of the wound healing areas were captured and measured with a Leica DM2500 image analysis system (Leica, Mannheim, Germany).

Cell migration assay

The cell migration assay was performed to analyze the effect of SPARCL1 on the migration of SKOV-3 cells. SKOV-3 cells (2.5×104 cells) were suspended in serum-free medium and seeded into the upper chamber of a transwell plate (Corning Costar, Rochester, NY, USA). RPMI-1640 medium containing 10% FBS was added in the lower chamber. The cells were allowed to migrate for 24 h at 37°C. Then, the non-migratory cells that remained in the upper chamber were removed, while the cells that had migrated to the lower chamber were fixed and stained with 0.1% crystal violet. Six randomly selected fields were photographed, and the cells per field of view were counted under a light microscope (at ×200 magnification).

Western blot analysis

Following treatment, the SKOV-3 cells were harvested and lysed in protein lysis buffer supplemented with a protease inhibitor (Beyotime Institute of Biotechnology). The total protein concentrations were determined using a bicinchoninic acid (BCA) protein assay kit (Bio-Rad Laboratories, Inc.). Equal amounts of protein were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred to a polyvinylidene fluoride (PVDF) membrane (EMD Millipore, Billerica, MA, USA). After blocking in 5% non-fat dry milk for 2 h at room temperature, the membranes were incubated overnight at 4°C with primary antibodies specific to GAPDH (1:8,000; Cell Signaling Technology, Inc., Danvers, MA, USA), SPARCL1 (1:1,000; Abcam), p-mitogen-activated protein kinase kinase (p-MEK; 1:1,000; Cell Signaling Technology, Inc.), t-MEK (1:1,000; Cell Signaling Technology, Inc.), p-extracellular signal-related kinase (p-ERK; 1:1,000; Cell Signaling Technology, Inc.), t-ERK (1:1,000; Cell Signaling Technology, Inc.) and MMP-2 (1:1,000; Cell Signaling Technology, Inc.). Then the membrane was incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies (1:1,000) for 2 h at room temperature. Immunoreactive bands were developed by enhanced chemiluminescence reagents (Pierce; Thermo Fisher Scientific, Inc.) and quantified using Bio-Rad Quantity One software (Bio-Rad Laboratories, Inc.).

Statistical analysis

All experiments were conducted at least three times. Data were presented as the mean ± standard deviation. The results between groups were statistically analyzed by Student's t-test or one-way analysis of variance with Bonferroni's post hoc test using SPSS 23.0 software (IBM Corp., Armonk, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

SPARCL1 was downregulated in clinical ovarian cancer specimens

The expression levels of SPARCL1 in ovarian cancer tissues and adjacent normal tissues was determined by IHC using SPARCL1 specific antibodies. As shown in Fig. 1A, SPARCL1 was predominantly localized to cytoplasm of the adjacent normal tissues, whereas there was none or low level staining observed in ovarian cancer tissues. After quantification we found that significantly lower levels of SPARCL1 were detected in ovarian cancer tissues than in adjacent normal tissues (P<0.05; Fig. 1).

SPARCL1 inhibited SKOV-3 cell proliferation

Western blotting analysis verified the successful up- or down-regulation of SPARCL1 expression levels in SKOV-3 cells after lentiviral transfection (Fig. 2). The results of the MTT assay revealed a significantly lower proliferation level in the SPARCL1 overexpression group compared with the other groups. After knock-down of SPARCL1, the proliferation rate increased significantly (Fig. 3A).

SPARCL1 inhibits migration of SKOV-3 cells

Relevant photographs of wound healing assay were taken immediately after induction of the wound and 24 h later on the following day. SPARCL1-overexpressing SKOV-3 cells had significantly lower levels of wound healing than those of the control group. In contrast, wound healing was significantly enhanced after SPARCL1 knockdown (Fig. 3B and D). In order to further examine the effect of SPARCL1 on human ovarian cancer cell migration, a transwell assay was performed. SPARCL1-overexpression or knockdown was initiated in SKOV-3 cells for 24 h. As shown in Fig. 3C and E, the migration of SKOV-3 cells was significantly inhibited by SPARCL1 overexpression. Furthermore, migration of SKOV-3 cells was significantly enhanced after SPARCL1 knockdown (Fig. 3C and E).

SPARCL1 inhibited the MEK/ERK signaling pathway

To investigate the underlying mechanism of action of SPARCL1 that affected the proliferation and migration of SKOV-3 cells, we investigated different key players of the MEK/ERK signaling pathway, because this pathway has a key regulator function in ovarian cancer cells. As shown in Fig. 4A, western blotting analysis showed that SPARCL1 overexpression led to a significantly reduced expression of activated MMP-2, while SPARCL1 knockdown led to increased expression levels of activated MMP-2. In addition, SPARCL1 overexpression significantly decreased the levels of p-MEK and p-ERK. In contrast, the expression of p-MEK and p-ERK were significantly upregulated in SPARCL1-knockdown cells (Fig. 4A-C).

To examine the mechanism by which SPARCL1 mediated the inhibition of MEK/ERK, a MEK1 specific inhibitor PD98059 (50 µmol/l), was used to inhibit MEK expression. As shown in Fig. 4C, PD98059 significantly inhibited the expression of matured MMP-2. In addition, the phosphorylation of MEK and ERK was blocked by the pretreatment of the cells with PD98059 (Fig. 4C). Furthermore, the results of the wound healing assay showed that the increased migration that was induced by SPARCL1 knockdown were also reduced after PD98059 exposure (Fig. 4D), suggesting that the inhibition effect of SPARCL1 on SKOV-3 cells may be mediated, at least partly, due to the downregulation of MEK/ERK signaling.

Discussion

Although previous studies have revealed a role of SPARCL1 in tumor suppression (59), it remains unknown whether SPARCL1 has any effect on ovarian cancer cell proliferation and migration and by which molecular mechanisms. The present study investigated the role of SPARCL1 in ovarian cancer. Our results show that the expression of SPARCL1 in ovarian cancer tissues was significantly lower than in the adjacent normal tissues. SPARCL1 overexpression significantly inhibited the proliferation and migration of SKOV-3 cells. Moreover, the tumor suppressing effect of SPARCL1 might be mediated by inhibiting the MEK/ERK pathway and downregulating the expression of MMP-2.

SPARCL1 was found to be downregulated in several human tumors, particularly in non-small cell lung cancer (NSCLC), pancreas and colon carcinomas, osteosarcoma, and prostate cancers (57,1316). Previous studies have demonstrated SPARCL1 to mediate cell detachment of cultured cells in vitro and suggested a putative role of SPARCL1 as tumor suppressor (6,7,16). Bendik et al (17) found that SPARCL1 was significantly downregulated in nine patients with NSCLC. In another study, Xiang et al (7) found that SPARCL1 inhibited the migration and invasion in a prostate cancer cell line both in vitro and in vivo, suggesting a putative role of SPARCL1 in suppressing prostate cancer metastasis. Naschberger et al (18) reported that SPARCL1 is of key importance for the modulation of the tumor microenvironment in colorectal carcinoma, by promoting stromal cell plasticity. Moreover, SPARCL1 was more strongly expressed in highly differentiated tumors than in lowly differentiated ones, which correlated significantly with patient prognosis (19). Similarly, in our study, IHC revealed lower SPARCL1 expression in ovarian cancer samples, suggesting that low SPARCL1 expression levels may be associated with a better prognosis. Overexpression of SPARCL1 inhibited the proliferation and migration of human ovarian cancer cells, indicating that SPARCL1 may inhibit the development and metastasis of ovarian cancer.

The MEK/ERK pathway is one of the key signaling pathways involved in cell survival and proliferation that transmits the signal by phosphorylation of a series of downstream molecules (2027). In squamous cell carcinoma cells, MEK/ERK signaling has been demonstrated to support cell survival by inducing the anti-apoptotic protein Bcl-2 (28). Previous studies have shown that alteration of the MEK/ERK pathway is also strongly implicated in ovarian cancer pathogenesis (2937). Al-Ayoubi et al (38) reported that signaling via the MEK/ERK pathway helps cells escape anoikis and maintain anchorage-independent growth in several ovarian cancer cell lines. Moreover, several studies have suggested that inhibition of the MEK/ERK pathway disrupts functions essential for ovarian cancer progression (29,35,37). Dang et al (39) reported that metformin in combination with cisplatin inhibits cell viability and induces apoptosis of human ovarian cancer cells by inactivating ERK. In another study, Chan et al (40) demonstrated that therapies targeting the ERK signaling pathway reduce the aggressiveness of ovarian cancer cells. In our study, SPARCL1 was found to inhibit the expression of p-MEK and p-ERK via the MEK/ERK pathway, which was then rescued by a MEK inhibitor. In line with previous studies, we found that inhibition of MEK/ERK was associated with ovarian cancer cell proliferation and migration. Thus, we conclude that the inhibition effect of SPARCL1 on SKOV-3 cells may be, at least partly, due to the downregulation of MEK/ERK signaling.

To our knowledge, this is the first study that investigates the effect of SPARCL1 on ovarian cancer cells. However, a key limitation of this study is that is an in vitro study. Thus, the results of the current study need to be validated by in vivo experiments in suitable animal models. Furthermore, there may be additional signal pathways involved apart from the MEK/ERK signaling. Further studies are required to determine possible alternative signal pathways that may also play a role in ovarian cancer progression.

In summary, our results show that SPARCL1 suppresses ovarian cancer cell proliferation and migration by downregulating signaling via the MEK/ERK pathway. These findings suggest that SPARCL1, a possible tumor suppressor, has potential as therapeutic target for developing novel therapeutic strategies in the prevention and intervention of ovarian cancers.

Acknowledgements

Not applicable.

Funding

The present study was financially supported by the Natural Science Foundation of Xinjiang Uygur Autonomous Region (grant no. 2017D01C409).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LL conceived and designed the present study. YM and YX performed the experiments, analyzed and interpreted the data, and were also the predominant contributors in the writing of the manuscript. YM made contributions to the interpretation of data and critically revised the manuscript for important intellectual content. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Medical Ethics Committee of the Third Affiliated Hospital, Xinjiang Medical University (Xinjiang, China). Signed written informed consent was obtained from all subjects.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen C, Wu J, Zhu P, Xu C and Yao L: Investigating isoquinoline derivatives for inhibition of inhibitor of apoptosis proteins for ovarian cancer treatment. Drug Des Devel Ther. 11:2697–2707. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Zhang W, Su J, Xu H, Yu S, Liu Y, Zhang Y, Sun L, Yue Y and Zhou X: Dicumarol inhibits PDK1 and targets multiple malignant behaviors of ovarian cancer cells. PLoS One. 12:e01796722017. View Article : Google Scholar : PubMed/NCBI

3 

Li Q, Gao JF and Qi BL: PDCD1 strengthens the sensitivity of ovarian cancer to cisplatin chemotherapy by promoting apoptosis. J BUON. 22:746–756. 2017.PubMed/NCBI

4 

Zhao Y, Cui L, Pan Y, Shao D, Zheng X, Zhang F, Zhang H, He K and Chen L: Berberine inhibits the chemotherapy-induced repopulation by suppressing the arachidonic acid metabolic pathway and phosphorylation of FAK in ovarian cancer. Cell Prolif. 50:e123932017. View Article : Google Scholar

5 

Zhao SJ, Jiang YQ, Xu NW, Li Q, Zhang Q, Wang SY, Li J, Wang YH, Zhang YL, Jiang SH, et al: SPARCL1 suppresses osteosarcoma metastasis and recruits macrophages by activation of canonical WNT/β-catenin signaling through stabilization of the WNT-receptor complex. Oncogene. 37:1049–1061. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Ye H, Wang WG, Cao J and Hu XC: SPARCL1 suppresses cell migration and invasion in renal cell carcinoma. Mol Med Rep. 16:7784–7790. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Xiang Y, Qiu Q, Jiang M, Jin R, Lehmann BD, Strand DW, Jovanovic B, DeGraff DJ, Zheng Y, Yousif DA, et al: Yi Y: SPARCL1 suppresses metastasis in prostate cancer. Mol Oncol. 7:1019–1030. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Wu DM, Shi J, Liu T, Deng SH, Han R and Xu Y: Integrated analysis reveals down-regulation of SPARCL1 is correlated with cervical cancer development and progression. Cancer Biomark. 21:355–365. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Liu X, Gao Y, Zhao B, Li X, Lu Y, Zhang J, Li D, Li L and Yin F: Discovery of microarray-identified genes associated with ovarian cancer progression. Int J Oncol. 46:2467–2478. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Jiang G, Yang D, Wang L, Zhang X, Xu H, Miao Y, Wang E and Zhang Y: A novel biomarker ARMc8 promotes the malignant progression of ovarian cancer. Hum Pathol. 46:1471–1479. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Josahkian JA, Saggioro FP, Vidotto T, Ventura HT, Candido Dos Reis FJ, de Sousa CB, Tiezzi DG, de Andrade JM, Koti M and Squire JA: Increased STAT1 expression in high grade serous ovarian cancer is associated with a better outcome. Int J Gynecol Cancer. 28:459–465. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Wei W, Li Y, Lv S, Zhang C and Tian Y: PARP-1 may be involved in angiogenesis in epithelial ovarian cancer. Oncol Lett. 12:4561–4567. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Cao F, Wang K, Zhu R, Hu YW, Fang WZ and Ding HZ: Clinicopathological significance of reduced SPARCL1 expression in human breast cancer. Asian Pac J Cancer Prev. 14:195–200. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Hurley PJ, Hughes RM, Simons BW, Huang J, Miller RM, Shinder B, Haffner MC, Esopi D, Kimura Y, Jabbari J, et al: Androgen-regulated SPARCL1 in the tumor microenvironment inhibits metastatic progression. Cancer Res. 75:4322–4334. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Hurley PJ, Marchionni L, Simons BW, Ross AE, Peskoe SB, Miller RM, Erho N, Vergara IA, Ghadessi M, Huang Z, et al: Secreted protein, acidic and rich in cysteine-like 1 (SPARCL1) is down regulated in aggressive prostate cancers and is prognostic for poor clinical outcome. Proc Natl Acad Sci USA. 109:14977–14982. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Jakharia A, Borkakoty B and Singh S: Expression of SPARC like protein 1 (SPARCL1), extracellular matrix-associated protein is down regulated in gastric adenocarcinoma. J Gastrointest Oncol. 7:278–283. 2016.PubMed/NCBI

17 

Bendik I, Schraml P and Ludwig CU: Characterization of MAST9/Hevin, a SPARC-like protein, that is down-regulated in non-small cell lung cancer. Cancer Res. 58:626–629. 1998.PubMed/NCBI

18 

Naschberger E, Liebl A, Schellerer VS, Schutz M, Britzen-Laurent N, Kolbel P, Schaal U, Haep L, Regensburger D, Wittmann T, et al: Matricellular protein SPARCL1 regulates tumor microenvironment-dependent endothelial cell heterogeneity in colorectal carcinoma. J Clin Invest. 126:4187–4204. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Zhang H, Widegren E, Wang DW and Sun XF: SPARCL1: A potential molecule associated with tumor diagnosis, progression and prognosis of colorectal cancer. Tumour Biol. 32:1225–1231. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Chi F, Wu R, Jin X, Jiang M and Zhu X: HER2 induces cell proliferation and invasion of non-small-cell lung cancer by upregulating COX-2 expression via MEK/ERK signaling pathway. Onco Targets Ther. 9:2709–2716. 2016.PubMed/NCBI

21 

Li K, Guo Q, Yang J, Chen H, Hu K, Zhao J, Zheng S, Pang X, Zhou S, Dang Y and Li L: FOXD3 is a tumor suppressor of colon cancer by inhibiting EGFR-Ras-Raf-MEK-ERK signal pathway. Oncotarget. 8:5048–5056. 2017.PubMed/NCBI

22 

Shen MJ, Xu LJ, Yang L, Tsai Y, Keng PC and Chen Y, Lee SO and Chen Y: Radiation alters PD-L1/NKG2D ligand levels in lung cancer cells and leads to immune escape from NK cell cytotoxicity via IL-6-MEK/Erk signaling pathway. Oncotarget. 8:80506–80520. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Wang J, Guo X, Xie C and Jiang J: KIF15 promotes pancreatic cancer proliferation via the MEK-ERK signalling pathway. Br J Cancer. 117:245–255. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Wang Y, Xiao H, Wu H, Yao C, He H, Wang C and Li W: G protein subunit alpha q regulates gastric cancer growth via the p53/p21 and MEK/ERK pathways. Oncol Rep. 37:1998–2006. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Yang K, Gao K, Hu G, Wen Y, Lin C and Li X: CTGF enhances resistance to 5-FU-mediating cell apoptosis through FAK/MEK/ERK signal pathway in colorectal cancer. Onco Targets Ther. 9:7285–7295. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Yang L, Shen M, Xu LJ, Yang X, Tsai Y, Keng PC, Chen Y and Lee SO: Enhancing NK cell-mediated cytotoxicity to cisplatin-resistant lung cancer cells via MEK/Erk signaling inhibition. Sci Rep. 7:79582017. View Article : Google Scholar : PubMed/NCBI

27 

Zhang C, Yu P, Zhu L, Zhao Q, Lu X and Bo S: Blockade of alpha7 nicotinic acetylcholine receptors inhibit nicotine-induced tumor growth and vimentin expression in non-small cell lung cancer through MEK/ERK signaling way. Oncol Rep. 38:3309–3318. 2017.PubMed/NCBI

28 

Shen X and Kramer RH: Adhesion-mediated squamous cell carcinoma survival through ligand-independent activation of epidermal growth factor receptor. Am J Pathol. 165:1315–1329. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Bai RX, Wang WP, Zhao PW and Li CB: Ghrelin attenuates the growth of HO-8910 ovarian cancer cells through the ERK pathway. Braz J Med Biol Res. 49:e50432016. View Article : Google Scholar

30 

Bartholomeusz C, Itamochi H, Nitta M, Saya H, Ginsberg MH and Ueno NT: Antitumor effect of E1A in ovarian cancer by cytoplasmic sequestration of activated ERK by PEA15. Oncogene. 25:79–90. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Chang MC, Chen CA, Chen PJ, Chiang YC, Chen YL, Mao TL, Lin HW, Lin Chiang WH and Cheng WF: Mesothelin enhances invasion of ovarian cancer by inducing MMP-7 through MAPK/ERK and JNK pathways. Biochem J. 442:293–302. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Fujisawa T, Joshi BH and Puri RK: IL-13 regulates cancer invasion and metastasis through IL-13Rα2 via ERK/AP-1 pathway in mouse model of human ovarian cancer. Int J Cancer. 131:344–356. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Ohta T, Isobe M, Takahashi T, Saitoh-Sekiguchi M, Motoyama T and Kurachi H: The Akt and ERK activation by platinum-based chemotherapy in ovarian cancer is associated with favorable patient outcome. Anticancer Res. 29:4639–4647. 2009.PubMed/NCBI

34 

Song H, Wei M, Liu W, Shen S, Li J and Wang L: Cisplatin induced apoptosis of ovarian cancer A2780s cells by activation of ERK/p53/PUMA signals. Histol Histopathol. 33:73–79. 2018.PubMed/NCBI

35 

Su S, Lin X, Ding N, Zhang H, Zhang Q, Ding Y, Hou X and Tian Y: Effects of PARP-1 inhibitor and ERK inhibitor on epithelial mesenchymal transitions of the ovarian cancer SKOV3 cells. Pharmacol Rep. 68:1225–1229. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Vergara D, Simeone P, Toraldo D, Del Boccio P, Vergaro V, Leporatti S, Pieragostino D, Tinelli A, De Domenico S, Alberti S, et al: Resveratrol downregulates Akt/GSK and ERK signalling pathways in OVCAR-3 ovarian cancer cells. Mol Biosyst. 8:1078–1087. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Wang W, Ren F, Wu Q, Jiang D, Li H and Shi H: MicroRNA-497 suppresses angiogenesis by targeting vascular endothelial growth factor A through the PI3K/AKT and MAPK/ERK pathways in ovarian cancer. Oncol Rep. 32:2127–2133. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Al-Ayoubi A, Tarcsafalvi A, Zheng H, Sakati W and Eblen ST: ERK activation and nuclear signaling induced by the loss of cell/matrix adhesion stimulates anchorage-independent growth of ovarian cancer cells. J Cell Biochem. 105:875–884. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Dang JH, Jin ZJ, Liu XJ, Hu D, Wang J, Luo Y and Li LL: Metformin in combination with cisplatin inhibits cell viability and induces apoptosis of human ovarian cancer cells by inactivating ERK 1/2. Oncol Lett. 14:7557–7564. 2017.PubMed/NCBI

40 

Chan DW, Hui WW, Cai PC, Liu MX, Yung MM, Mak CS, Leung TH, Chan KK and Ngan HY: Targeting GRB7/ERK/FOXM1 signaling pathway impairs aggressiveness of ovarian cancer cells. PLoS One. 7:e525782012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 16 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
Ma Y, Xu Y and Li L: SPARCL1 suppresses the proliferation and migration of human ovarian cancer cells via the MEK/ERK signaling. Exp Ther Med 16: 3195-3201, 2018
APA
Ma, Y., Xu, Y., & Li, L. (2018). SPARCL1 suppresses the proliferation and migration of human ovarian cancer cells via the MEK/ERK signaling. Experimental and Therapeutic Medicine, 16, 3195-3201. https://doi.org/10.3892/etm.2018.6575
MLA
Ma, Y., Xu, Y., Li, L."SPARCL1 suppresses the proliferation and migration of human ovarian cancer cells via the MEK/ERK signaling". Experimental and Therapeutic Medicine 16.4 (2018): 3195-3201.
Chicago
Ma, Y., Xu, Y., Li, L."SPARCL1 suppresses the proliferation and migration of human ovarian cancer cells via the MEK/ERK signaling". Experimental and Therapeutic Medicine 16, no. 4 (2018): 3195-3201. https://doi.org/10.3892/etm.2018.6575