MCM7 amplification and overexpression promote cell proliferation, colony formation and migration in esophageal squamous cell carcinoma by activating the AKT1/mTOR signaling pathway

  • Authors:
    • Yun-Tan Qiu
    • Wen-Jun Wang
    • Bing Zhang
    • Li-Li Mei
    • Zhi-Zhou Shi
  • View Affiliations

  • Published online on: May 2, 2017     https://doi.org/10.3892/or.2017.5614
  • Pages: 3590-3596
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The roles and mechanisms of mini-chromosome maintenance complex component 7 (MCM7) amplification and overexpression in esophageal carcinogenesis were investigated. By analyzing the TCGA datasets, we found that MCM7 was amplified in approximately 12% of esophageal squamous cell carcinomas (ESCCs), and in more than 4% of head and neck squamous cell carcinomas and stomach carcinomas. Overexpression of MCM7 was further verified in three independent GEO datasets of esophageal cancer. Knockdown of MCM7 using two siRNAs significantly inhibited cell proliferation, colony formation and migration of KYSE510 and EC9706 cells in vitro. Noteworthy, we further found that silencing of MCM7 suppressed the phosphorylation of AKT1 and mTOR both in KYSE510 and EC9706 cells, and reduced the cell cycle regulatory proteins cyclin D1, cyclin E2 and CDK2. Taken together, our findings suggested that MCM7 promoted tumor cell proliferation, colony formation and migration of ESCC cells via activating AKT1/mTOR signaling pathway.

Introduction

Esophageal cancer is the sixth leading cause of cancer-related death in the world and the fourth leading cause of cancer-related death in China (1). There are two types of esophageal cancer, squamous cell carcinoma (SCC) and adenocarcinoma (AC), and most Chinese esophageal cancers are ESCCs (2). Although diagnostic methods and cancer treatments have improved in recent years, the prognosis is still poor because of widespread lymph node metastasis and relatively frequent distant metastasis. Therefore, understanding the mechanism of esophageal carcinogenesis will lay the foundations for improving clinical management and outcomes.

Mini-chromosome maintenance protein complex (MCM) is a eukaryotic DNA helicase complex required for initiation of DNA replication. The MCM proteins include six members, MCM2 to MCM7, and are considered as molecular markers of proliferation in several types of cancer (36). MCM7 is a critical component of DNA replication licensing complex, and is overexpressed in multiple human malignancies including hepatocellular carcinoma, head and neck squamous cell carcinoma, prostate carcinoma and esophageal squamous cell carcinoma (710). Kim et al found that upregulation of MCM7 was associated with cisplatin resistance in bladder cancer (11). In liver cancer MHCC-97 cells, silence of MCM7 dramatically reduced the cell proliferation, migration, invasion and increased the apoptotic cells (12).

Our previous and other studies found that MCM7 was amplified and overexpressed in ESCC, and overexpression of MCM7 was significantly linked with poor prognosis (7,13,14), however the roles and mechanisms of MCM7 amplification and overexpression in ESCC were largely unclear. In the present study, we revealed that MCM7 promoted tumor cell proliferation, colony formation and migration of ESCC cells via inhibiting the AKT1/mTOR signaling pathway.

Materials and methods

TCGA and GEO datasets

Genomic and expression data of MCM7 are publically available from the Cancer Genome Atlas and the NCBI Gene Expression Omnibus. The copy number alterations and mutations of TCGA datasets were analyzed by Cbioportal (www.cbioportal.org). For GSE datasets of GSE20347, GSE38129 and GSE29001, the mRNA expression levels are detected by microarray, and the difference of MCM7 between ESCC tissues and paracancerous tissues was analyzed using the paired t-test by SPSS 19.

Cell culture

The human esophageal cancer cell lines were supplied by Peking Union Medical College and Chinese Academy of Medical Sciences. All cell lines were cultured in RPMI-1640 medium with 10% fetal bovine serum, 100 U/ml penicillin and 100 g/ml streptomycin at 37°C in a humidified incubator containing 5% carbon dioxide.

siRNAs and transfection

The synthetic negative control siRNA, MCM7 siRNA-1 and MCM7 siRNA-2 were purchased from Shanghai Gene Pharma Co. Ltd. The ESCC cell lines were transiently transfected using Lipofectamine® 2000 Transfection Reagent from Invitrogen according to the manufacturer's protocol. The sequences of negative control siRNA, MCM7 siRNA-1 and MCM7 siRNA-2 were as follows: Negative control siRNA: 5′-UUCUCCGAACGUGUCACGUTT-3′; MCM7 siRNA-1: 5′-ATCGGATTGTGAAGATGAA-3′; MCM7 siRNA-2: 5′-AAGAUGUCCUGGACGUUUACA-3′.

Total RNA extraction and real-time PCR assay

Total RNA was isolated from cancer cells using the RNeasy mini kit as described by the manufacturer (Qiagen, Hilden, Germany) and used for Real-time PCR assay.

Real-time PCR was used to detect the mRNA expression levels of cyclin D1, cyclin E2 and CDK2. The PCR reactions were performed in a total volume of 20 µl, including 10 µl of 2X Power SYBR® Green PCR Master Mix (Applied Biosystems, Warrington, UK), 2 µl of cDNA (5 ng/µl) and 1 µl of primer mix (10 µM each). PCR amplification and detection were performed in a LightCycler 480 II (Roche Applied Science) as follows: an initial denaturation at 95°C for 10 min; 40 cycles at 95°C for 15 sec and 60°C for 1 min. The relative gene expression was calculated using the comparative CT method. The gene expression of the target gene were normalized to an endogenous reference (GAPDH), and relative to the calibrator were given by the formula 2-∆∆Ct. ∆CT was calculated by subtracting the average GAPDH CT from the average CT of the gene of interest. The ratio defines the level of relative expression of the target gene to that of GAPDH. The primers were as follows: Cyclin D1 forward primer, 5′-ACGCTTACCTCAACCATCCTG-3′; Cyclin D1 reverse primer, 5′-GGCCTCTCGATACACACAACA-3′; Cyclin E2 forward primer, 5′-GCCCGGCCTATATATTGGGTT-3′; Cyclin E2 reverse primer, 5′-AACGGCTACTTCGTCTTGACA-3′; CDK2 forward primer, 5′-TCTTTGCTGAGATGGTGACTCG-3′; CDK2 reverse primer, 5′-TCTTCATCCAGGGGAGGTACA-3′; GAPDH forward primer, 5′-AAATCCCATCACCATCTTCCAG-3′; GAPDH reverse primer, 5′-GAGTCCTTCCACGATACCAAAGTTG-3′.

Western blot assay

Cells were lysed in the lysis buffer (20 mM Tris, 2 mM EDTA, 50 mM 2-mecaptoethanol, 10% glycerol, pH 7.4). The homogenates were placed on ice for 30 min and centrifuged at 12,000 × g for 15 min at 4°C. Afterward, the protein concentrations of the lysates were determined using a Protein Assay kit (Bio-Rad, Richmond, CA, USA). Equal amounts of total proteins were loaded onto a 10% gradient polyacrylamide gel, electrophoretically transferred to polyvinylidene difluoride membrane, and then blocked with 10% non-fat milk for 2 h at room temperature. The membranes were incubated with specific primary antibodies overnight at 4°C and probed with corresponding secondary antibodies for 1 h at room temperature. The protein bands were visualized using ECL Blotting Detection Reagents (Applygen, Beijing, China). The primary antibodies were as follows: MCM7 (1:1000; Santa Cruz Biotechnology, Santa Cruz, CA, USA), p-AKT1 (1:1000; Santa Cruz Biotechnology), AKT1 (1:1000; Santa Cruz Biotechnology), p-mTOR (1:1000; Santa Cruz Biotechnology), mTOR (1:1000; Santa Cruz Biotechnology) and β-actin (1:5000; Santa Cruz Biotechnology).

Cell proliferation assay

The Cell Counting Kit-8 (Dojindo Laboratories, Kumamoto, Japan) was performed to quantify the proliferation of KYSE510 and EC9706 cells. Cells were cultured at 1000/well in 96-well plates. After incubated for 24, 48, 72, 84, 96, or 120 h, 10 µl of CCK-8 was added to each well and incubated for 1 h. The absorbance of each well was read at 450 nm. Three independent experiments were performed.

Colony formation assay

For each group, 5000 cells were plated in 6-well plate. After cultured for 10 days, the cells were washed with PBS, fixed with methanol and 0.1% crystal violet. Then, the colonies were counted and photographed. Three independent experiments were performed.

Transwell assay

The migration assay was performed on Transwell plates. For cell migration assay, 1×105 cells were seeded on a polycarbonate membrane insert in a Transwell apparatus (Costar, Cambridge, MA, USA) and cultured in RPMI-1640 without serum. RPMI-1640 containing 20% fetal bovine serum was added to the lower chamber. After incubation for 24 h at 37°C in a CO2 incubator, the insert was washed with PBS, and cells on the top surface of the insert were removed by wiping with a cotton swab. Cells that migrated to the bottom surface of the insert were fixed with methanol, stained with 0.4% crystal violet, and counted in five random fields at ×200.

Statistical analysis

The data were analyzed by Student's t-test and one-way analysis of variance using the SPSS and Graphpad Prism 5.0. P<0.05 was considered to indicate a statistical significant difference.

Results

MCM7 is amplified and overexpressed in many types of cancer including ESCC

The datasets of The Cancer Genome Atlas (TCGA) showed that MCM7 was amplified in approximately 12% of ESCCs, and in >4% of head and neck squamous cell carcinomas and stomach carcinomas, and the amplification was the dominant form of changes in DNA level; however in diffuse large B-cell lymphoma (DLBC), mutation was more frequent than amplification (Fig. 1). MCM7 was overexpressed in ESCC tissues in the datasets of GSE20347, GSE38129 and GSE29001 (Fig. 2).

Knockdown of MCM7 suppresses the proliferation, colony formation and migration of ESCC cells

In order to explore the tumorigenic roles and mechanisms of MCM7 in esophageal carcinogenesis, we selected cell lines KYSE510 and EC9706 with higher MCM7 expression for further study.

siRNAs were used to knock down the MCM7 expression in KYSE510 and EC9706 cells, and the RNAi efficiency was determined by western blot assay (Fig. 3A and B). Using CCK-8 proliferation assay, we found that silence of MCM7 significantly inhibited cell proliferation compared with non-silencing group, and there was significant suppression at days 4, 5, 6 and 7 both in KYSE510 and EC9706 cells (Fig. 4A and B). In colony formation assay, we observed that colony numbers were significantly lower in MCM7 siRNA-1 and siRNA-2 transfected cells than in the control group both in KYSE510 and EC9706 cells (Fig. 4C and D). By Transwell assay, we revealed that silence of MCM7 significantly inhibited the migration of KYSE510 and EC9706 cells (Fig. 4E and F).

Knockdown of MCM7 suppressed the AKT1/mTOR signaling pathway in ESCC

Many studies showed that activated AKT1/mTOR signaling pathway promoted cancer cell proliferation, epithelial-mesenchymal transition (EMT), tumor metastasis and invasion (1520). Noteworthy, our study revealed that silencing MCM7 significantly inhibited the phosphorylation of AKT1 and mTOR (Fig. 5). In addition, knockdown of MCM7 reduced the cell cycle regulatory genes cyclin D1, cyclin E2 and CDK2 in mRNA expression levels (Fig. 6). These results indicated that MCM7 amplification and overexpression promoted cell proliferation, colony formation and migration via activating the AKT1/mTOR signaling pathway.

Discussion

Genomic aberrations can contribute to carcinogenesis and tumor progression. In the past decades, the understanding of molecular pathogenesis of ESCC has developed, but is still limited. Thus, elucidation of the mechanism of esophageal carcinogenesis was very important for tumor diagnosis and therapy. Recently, it was shown that MCM proteins were overexpressed in several types of tumors, and high expression levels of MCM proteins including MCM2, MCM3 and MCM7 were positively correlated with Ki-67 positive staining in Laryngeal squamous cell carcinoma (LSCC) (5).

MCM7 is a critical DNA replication licensing factor in both yeast and xenopus oocytes, and serves as a co-transcriptional and co-translational enhancing factor of androgen receptor, which regulates cell growth and proliferation (21). Increased MCM7 expression is common in various human cancers including esophageal squamous cell carcinoma, prostate cancer and pancreatic cancer (7,10,22,23), and MCM7 has been considered as a tumorigenesis-related gene (24). In pituitary adenoma, diffuse-type primary gastric adenocarcinoma and colorectal cancer, the patients with MCM7 overexpression had a shorter recurrence/progression-free survival respectively (25,26). MCM7 promotes tumor cell proliferation and invasion in papillary urothelial neoplasia and liver cancer (27), and depletion of MCM7 inhibits glioblastoma multiforme tumor growth in vivo (28). In ESCC, G9a and MCM7 overexpression levels were correlated with poor prognosis (7), however, the roles and mechanisms of MCM7 amplification and overexpression in ESCC were largely unknown. Our results showed that MCM7 promoted tumor cell proliferation, colony formation and migration of ESCC cells.

AKT, a serine/threonine kinase, has a wide tissue distribution and regulates many processes including cell metabolism, proliferation, survival and tumor growth (29). Notably, AKT has been considered as a critical oncogene, and it can activate downstream signaling pathways through phosphorylation of a plethora of AKT substrates (30). In recent studies, the phosphorylation of AKT was shown correlated with poor prognosis in ESCC (31,32). AKT was responsible for the cisplatin resistance, and could promote metastasis of ESCC by targeting epithelial-mesenchymal transition (3335). mTOR activation was able to also prcomote the cell proliferation and tumor progression of ESCC (36,37). However, the direct relationship between AKT/mTOR and MCM7 has not been reported. Our findings further revealed that knockdown of MCM7 significantly inhibited the phosphorylation of AKT and mTOR.

In summary, our data revealed that MCM7 promoted tumor cell proliferation, colony formation and migration via activating the AKT1/mTOR signaling pathway. Future studies should focus on how MCM7 regulates the AKT1/mTOR signaling pathway, and explore the antitumor activity of MCM7 silencing in an animal model.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (no. 81460425) and the Yunnan Provincial Research Foundation for Basic Research, China (no. 2013FD012) and Foundation for the Talents of Kunming University of Science and Technology (no. KKSY201226099).

References

1 

Chen W, Zheng R, Zuo T, Zeng H, Zhang S and He J: National cancer incidence and mortality in China, 2012. Chin J Cancer Res. 28:1–11. 2016. View Article : Google Scholar : PubMed/NCBI

2 

He YT, Hou J, Chen ZF, Qiao CY, Song GH, Meng FS, Jin HX and Chen C: Decrease in the esophageal cancer incidence rate in mountainous but not level parts of Cixian County, China, over 29 years. Asian Pac J Cancer Prev. 6:510–514. 2005.PubMed/NCBI

3 

Chang CC, Huang CC, Yang SH, Chien CC, Lee CL and Huang CJ: Data on clinical significance of GAS2 in colorectal cancer cells. Data Brief. 8:82–86. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Juríková M, Danihel Ľ, Polák Š and Varga I: Ki67, PCNA, and MCM proteins: Markers of proliferation in the diagnosis of breast cancer. Acta Histochem. 118:544–552. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Nowinska K, Chmielewska M, Piotrowska A, Pula B, Pastuszewski W, Krecicki T, Podhorska-Okołow M, Zabel M and Dziegiel P: Correlation between levels of expression of minichromosome maintenance proteins, Ki-67 proliferation antigen and metallothionein I/II in laryngeal squamous cell cancer. Int J Oncol. 48:635–645. 2016.PubMed/NCBI

6 

Gann PH, Deaton RJ, Rueter EE, van Breemen RB, Nonn L, Macias V, Han M and Ananthanarayanan V: A phase II randomized trial of lycopene-rich tomato extract among men with high-grade prostatic intraepithelial neoplasia. Nutr Cancer. 67:1104–1112. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Zhong X, Chen X, Guan X, Zhang H, Ma Y, Zhang S, Wang E, Zhang L and Han Y: Overexpression of G9a and MCM7 in oesophageal squamous cell carcinoma is associated with poor prognosis. Histopathology. 66:192–200. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Zhou YM, Zhang XF, Cao L, Li B, Sui CJ, Li YM and Yin ZF: MCM7 expression predicts post-operative prognosis for hepatocellular carcinoma. Liver Int. 32:1505–1509. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Honeycutt KA, Chen Z, Koster MI, Miers M, Nuchtern J, Hicks J, Roop DR and Shohet JM: Deregulated minichromosomal maintenance protein MCM7 contributes to oncogene driven tumorigenesis. Oncogene. 25:4027–4032. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Ren B, Yu G, Tseng GC, Cieply K, Gavel T, Nelson J, Michalopoulos G, Yu YP and Luo JH: MCM7 amplification and overexpression are associated with prostate cancer progression. Oncogene. 25:1090–1098. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Kim SH, Ho JN, Jin H, Lee SC, Lee SE, Hong SK, Lee JW, Lee ES and Byun SS: Upregulated expression of BCL2, MCM7, and CCNE1 indicate cisplatin-resistance in the set of two human bladder cancer cell lines: T24 cisplatin sensitive and T24R2 cisplatin resistant bladder cancer cell lines. Investig Clin Urol. 57:63–72. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Liu J, Tian L, Chen BA and Xia JR: Biological effects of lentivirus-mediated silencing of minichromosome maintenance protein 7 with shRNA on the liver cancer MHCC-97H cells. Int J Clin Exp Med. 8:8433–8441. 2015.PubMed/NCBI

13 

Kan T, Sato F, Ito T, Matsumura N, David S, Cheng Y, Agarwal R, Paun BC, Jin Z, Olaru AV, et al: The miR-106b-25 polycistron, activated by genomic amplification, functions as an oncogene by suppressing p21 and Bim. Gastroenterology. 136:1689–1700. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Shi ZZ, Shang L, Jiang YY, Hao JJ, Zhang Y, Zhang TT, Lin DC, Liu SG, Wang BS, Gong T, et al: Consistent and differential genetic aberrations between esophageal dysplasia and squamous cell carcinoma detected by array comparative genomic hybridization. Clin Cancer Res. 19:5867–5878. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Leu WJ, Swain Sh P, Chan SH, Hsu JL, Liu SP, Chan ML, Yu CC, Hsu LC, Chou YL, Chang WL, et al: Non-immunosuppressive triazole-based small molecule induces anticancer activity against human hormone-refractory prostate cancers: The role in inhibition of PI3K/AKT/mTOR and c-Myc signaling pathways. Oncotarget. 7:76995–77009. 2016.PubMed/NCBI

16 

Liu L, Pan Y, Song Y, Su X, Ke R, Yang L, Gao L and Li M: Activation of AMPK α2 inhibits airway smooth muscle cells proliferation. Eur J Pharmacol. 791:235–243. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Moravčík R, Stebelová K, Boháč A and Zeman M: Inhibition of VEGF mediated post receptor signalling pathways by recently developed tyrosine kinase inhibitor in comparison with sunitinib. Gen Physiol Biophys. 35:511–514. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Divine LM, Nguyen MR, Meller E, Desai RA, Arif B, Rankin EB, Bligard KH, Meyerson C, Hagemann IS, Massad M, et al: AXL modulates extracellular matrix protein expression and is essential for invasion and metastasis in endometrial cancer. Oncotarget. 7:77291–77305. 2016.PubMed/NCBI

19 

Yang Y, Gao Z, Ma Y, Teng H, Liu Z, Wei H, Lu Y, Cheng X, Hou L and Zou X: Fucoidan inhibits lymphangiogenesis by downregulating the expression of VEGFR3 and PROX1 in human lymphatic endothelial cells. Oncotarget. 7:38025–38035. 2016.PubMed/NCBI

20 

Wei L, Li K, Pang X, Guo B, Su M, Huang Y, Wang N, Ji F, Zhong C, Yang J, et al: Leptin promotes epithelial-mesenchymal transition of breast cancer via the upregulation of pyruvate kinase M2. J Exp Clin Cancer Res. 35:1662016. View Article : Google Scholar : PubMed/NCBI

21 

Shi YK, Yu YP, Zhu ZH, Han YC, Ren B, Nelson JB and Luo JH: MCM7 interacts with androgen receptor. Am J Pathol. 173:1758–1767. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Lau KM, Chan QK, Pang JC, Li KK, Yeung WW, Chung NY, Lui PC, Tam YS, Li HM, Zhou L, et al: Minichromosome maintenance proteins 2, 3 and 7 in medulloblastoma: Overexpression and involvement in regulation of cell migration and invasion. Oncogene. 29:5475–5489. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Liang JW, Shi ZZ, Shen TY, Che X, Wang Z, Shi SS, Xu X, Cai Y, Zhao P, Wang CF, et al: Identification of genomic alterations in pancreatic cancer using array-based comparative genomic hybridization. PLoS One. 9:e1146162014. View Article : Google Scholar : PubMed/NCBI

24 

Huber AR, Tan D, Sun J, Dean D, Wu T and Zhou Z: High expression of carbonic anhydrase IX is significantly associated with glandular lesions in gastroesophageal junction and with tumorigenesis markers BMI1, MCM4 and MCM7. BMC Gastroenterol. 15:802015. View Article : Google Scholar : PubMed/NCBI

25 

Coli A, Asa SL, Fadda G, Scannone D, Chiloiro S, De Marinis L, Lauretti L, Ranelletti FO and Lauriola L: Minichromosome maintenance protein 7 as prognostic marker of tumor aggressiveness in pituitary adenoma patients. Eur J Endocrinol. 174:307–314. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Ishibashi Y, Kinugasa T, Akagi Y, Ohchi T, Gotanda Y, Tanaka N, Fujino S, Yuge K, Kibe S, Yoshida N, et al: Minichromosome maintenance protein 7 is a risk factor for recurrence in patients with Dukes C colorectal cancer. Anticancer Res. 34:4569–4575. 2014.PubMed/NCBI

27 

Guan B, Wang X, Yang J, Zhou C and Meng Y: Minichromosome maintenance complex component 7 has an important role in the invasion of papillary urothelial neoplasia. Oncol Lett. 10:946–950. 2015.PubMed/NCBI

28 

Erkan EP, Ströbel T, Lewandrowski G, Tannous B, Madlener S, Czech T, Saydam N and Saydam O: Depletion of minichromosome maintenance protein 7 inhibits glioblastoma multiforme tumor growth in vivo. Oncogene. 33:4778–4785. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Vivanco I and Sawyers CL: The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2:489–501. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Hers I, Vincent EE and Tavaré JM: Akt signalling in health and disease. Cell Signal. 23:1515–1527. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Zhu Z, Yu W, Fu X, Sun M, Wei Q, Li D, Chen H, Xiang J, Li H, Zhang Y, et al: Phosphorylated AKT1 is associated with poor prognosis in esophageal squamous cell carcinoma. J Exp Clin Cancer Res. 34:952015. View Article : Google Scholar : PubMed/NCBI

32 

Kircher DA, Arave RA, Cho JH and Holmen SL: Melanoma metastases caught in the AKT. Mol Cell Oncol. 3:e11285162016. View Article : Google Scholar : PubMed/NCBI

33 

Shan B, Man H, Liu J, Wang L, Zhu T, Ma M, Xv Z, Chen X, Yang X and Li P: TIM-3 promotes the metastasis of esophageal squamous cell carcinoma by targeting epithelial-mesenchymal transition via the Akt/GSK-3β/Snail signaling pathway. Oncol Rep. 36:1551–1561. 2016.PubMed/NCBI

34 

Liu T, Li R, Zhao H, Deng J, Long Y, Shuai MT, Li Q, Gu H, Chen YQ and Leng AM: eIF4E promotes tumorigenesis and modulates chemosensitivity to cisplatin in esophageal squamous cell carcinoma. Oncotarget. 7:66851–66864. 2016.PubMed/NCBI

35 

Li DJ, Shi M and Wang Z: RUNX3 reverses cisplatin resistance in esophageal squamous cell carcinoma via suppression of the protein kinase B pathway. Thorac Cancer. 7:570–580. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Ji YM, Zhou XF, Zhang J, Zheng X, Li SB, Wei ZQ, Liu T, Cheng DL, Liu P, Song K, et al: DEPTOR suppresses the progression of esophageal squamous cell carcinoma and predicts poor prognosis. Oncotarget. 7:14188–14198. 2016.PubMed/NCBI

37 

Zhang W, Lei C, Fan J and Wang J: miR-18a promotes cell proliferation of esophageal squamous cell carcinoma cells by increasing cylin D1 via regulating PTEN-PI3K-AKT-mTOR signaling axis. Biochem Biophys Res Commun. 477:144–149. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2017
Volume 37 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qiu Y, Wang W, Zhang B, Mei L and Shi Z: MCM7 amplification and overexpression promote cell proliferation, colony formation and migration in esophageal squamous cell carcinoma by activating the AKT1/mTOR signaling pathway. Oncol Rep 37: 3590-3596, 2017
APA
Qiu, Y., Wang, W., Zhang, B., Mei, L., & Shi, Z. (2017). MCM7 amplification and overexpression promote cell proliferation, colony formation and migration in esophageal squamous cell carcinoma by activating the AKT1/mTOR signaling pathway. Oncology Reports, 37, 3590-3596. https://doi.org/10.3892/or.2017.5614
MLA
Qiu, Y., Wang, W., Zhang, B., Mei, L., Shi, Z."MCM7 amplification and overexpression promote cell proliferation, colony formation and migration in esophageal squamous cell carcinoma by activating the AKT1/mTOR signaling pathway". Oncology Reports 37.6 (2017): 3590-3596.
Chicago
Qiu, Y., Wang, W., Zhang, B., Mei, L., Shi, Z."MCM7 amplification and overexpression promote cell proliferation, colony formation and migration in esophageal squamous cell carcinoma by activating the AKT1/mTOR signaling pathway". Oncology Reports 37, no. 6 (2017): 3590-3596. https://doi.org/10.3892/or.2017.5614