Downregulation of Notch1 inhibits the invasion and metastasis of human gastric cancer cells SGC7901 and MKN74 in vitro through PTEN activation and dephosphorylation of Akt and FAK

  • Authors:
    • Xue‑Song Zhang
    • Yan‑Hua Hu
    • Hong‑Yu Gao
    • Xiu‑Wen Lan
    • Ying‑Wei Xue
  • View Affiliations

  • Published online on: June 15, 2017     https://doi.org/10.3892/mmr.2017.6791
  • Pages: 2318-2324
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Migration and invasion are both vital causes of mortality in patients with gastric cancer. Therefore, the inhibition of these tumour cell processes is of great importance in gastric cancer therapy. Activation of Notch has been reported in many cancers. The critical role of Notch and its regulation in tumourigenesis has been noted. Although the studies on Notch in the field of cancer have been performed extensively, the role of Notch1 signalling in gastric cancer requires further study. Inactivation of PTEN has been observed in the development of many malignant tumors, and loss of PTEN function has been implicated in tumorigenic processes. Notch acts as an upstream signalling pathway that regulates PTEN activities. However, the effect of Notch on invasion and metastasis in gastric cancer and the regulation of PTEN during this process remain poorly understood. In the present study, small interfering RNA (siRNA) was used to knock down Notch1 expression in gastric cancer cell lines SGC7901 and MKN74. The mRNA and protein expression of Notch1, PTEN, Akt and FAK were measured upon depletion of Notch1. phospho‑PTEN, phospho‑Akt and phospho‑FAK expression were measured using western blot analysis. Migration and invasion assays were also used after Notch1 depletion. Our results showed that the knockdown of Notch1 leads to the inhibition of cell invasion and metastasis of human gastric cancer cells SCG7901 and MKN74 in vitro. Compared to control and mock groups, PTEN activities were significantly promoted following depletion of Notch1, and the expression of Phospho‑Akt and Phospho‑FAK were downregulated. Taken together, our findings suggest that Notch1 could be used as a therapeutic target to inhibit cell invasion and migration in gastric cancer.

Introduction

Gastric cancer is one of the most malignant, aggressive and common cancers worldwide (1). According to recent statistics, the yearly estimated number of new gastric cancer cases was 26,370, and the yearly estimated number of gastric cancer-related deaths was 10,730 in the USA alone (2). These numbers were much higher in China: 679,100 and 498,000, respectively (3). Although the diagnosis and management of gastric cancer have greatly improved, this disease still accounts for 10% of all deaths caused by malignant tumors annually (4,5). The primary curative treatment of gastric cancer is surgical resection. However, Chinese gastric cancer patients are often diagnosed at advanced stages, making surgery much more difficult and less effective (6). Because advanced stages of gastric cancer are generally associated with greater invasion and metastasis, studies have suggested that inhibition of cell signalling pathways could greatly affect the invasion and metastasis of gastric cancer cells (712). Therefore, the new prevention strategy of targeting cell signalling pathways of invasion and metastasis in gastric cancer will likely be of great clinical value.

PTEN, which is also known as TGF-β-regulated and epithelial cell enriched phosphatase (TEP1) or muted in multiple advanced cancers (MMAC1), was isolated and identified by three laboratories in 1997 (1315). As a tumor suppressor gene, its protein is ubiquitously expressed among human cells. PTEN can function as a phosphatase to dephosphorylate phosphatidylinositol (3,4,5)-trisphosphate (PIP3) into the biphosphate product PIP2 (16). PIP3 is a primary activator of Akt, and dephosphorylation of PIP3 by PTEN results in inhibition of Akt signalling, which is critical in various cancer cellular functions, including cell transcription, proliferation, metastasis and invasion (16). PTEN also functions as a protein phosphatase by dephosphorylating FAK, restricting cancer cell invasion and metastasis (17,18). Studies have suggested that upstream signalling regulates invasion and metastasis of cancer cells through regulation of PTEN (19,20).

Notch is vitally important in controlling cell fate. The critical roles of Notch in tumorigenesis have been reported in many malignant tumors (21,22). Studies have indicated that Notch signalling has variable effects on different cancer cells. Notch signalling can either act as a tumor suppressor or tumor promoter (2325). The function of Notch signalling in gastric cancer, especially its effect on invasion and metastasis, remains unclear. Evidence has shown that Notch signalling regulates PTEN in cancer cells (12,26,27). However, the relationships between Notch and PTEN in gastric cancer cells require further research.

The aim of this study was to investigate the role and mechanism of the Notch1 signalling pathway on cell invasion and metastasis and possible downstream regulation during this process in gastric cancer cells in vitro.

In this study we determined that downregulation of Notch1 signalling using siRNA inhibits invasion and metastasis of gastric cancer cell lines SGC7901 and MKN74 in vitro. PTEN activation and decreased expression of phosphorylated forms of FAK and Akt was also observed after Notch1 depletion. Our data suggest that the Notch1 signalling pathway may provide an effective treatment in gastric cancer patients.

Materials and methods

Cell culture

The gastric cell line SGC7901 was kindly provided by the Second Affiliated Hospital of Harbin Medical University. The gastric cell line MKN74 was obtained from Harbin Medical University Cancer Hospital. Both cell lines were cultured in high glucose Dulbecco's modified Eagle's medium (DMEM; GE Healthcare Life Sciences; Hyclone, Logan, UT, USA) containing 10% foetal bovine serum (FBS; Biowest SAS, Nuaillé, France). All cells were incubated in 5% CO2 and 37°C in a humidified chamber.

RNA extraction and quantitative PCR

Total RNA was harvested from cell lines using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions. Reverse transcription were performed using the Golden 1st cDNA Synthesis kit (HaiGene, Harbin, China). Quantitative real-time PCR was performed with Mini-Opticon2 (MJ) by using the Golden HS SYBR-Green qPCR Mix (HaiGene). β-actin was used as an internal control and β-actin qPCR primer was obtained from HaiGene. The specific primers for each gene were synthesized by HaiGene. Specific primer sequences used were as follows: Notch1 forward, 5′-TCGGAGTGTGTATGCCAAGAG-3′ and reverse, 5′-TGATGCCTACATTTCAAGAACGG-3′; PTEN forward, 5′-GTGTGGAATGAAGTGAGGCTTG-3′ and reverse, 5′-TTGGACAACTGGATAGAGTAGGC-3′; Akt forward, 5′-CCATCACACCACCTGACCAAG-3′ and reverse, 5′-CGCCTCTCCATCCCTCCAAG-3′; FAK forward, 5′-GAGATTGAGATGGCACAGAAGC-3′ and reverse, 5′-TGAGCAGCAGTCAGCATTTG-3′. Specificity of amplification products was determined by melting curve analysis. Independent experiments were done in triplicate. The 2−ΔΔCt was presented as the relative expression of the gene expression.

Antibodies and reagents

The following primary antibodies were purchased from Cell Signaling Technology (CST; Danvers, MA, USA): Notch1, PTEN, Akt, FAK, phospho-PTEN (Ser380/Thr382/383), phospho-Akt (Ser473) and phospho-FAK (Tyr397). β-actin and all secondary antibodies were provided by Santa Cruz Biotechnology (Dallas, Texas, USA). Lipofectamine RNAiMAX, Notch1 small interfering RNA (siRNA), control siRNA, and related chemicals were purchased from Invitrogen.

siRNA transfection

The putative Notch1 candidate sequences and the control sequence were designed and provided by Invitrogen. The siRNA sequences are as follows: Sequence 1 forward, 5′-CCGCCUUUGUGCUUCUGUUCUUCGU-3′ and reverse, 5′-ACGAAGAACAGAAGCACAAAGGCGG-3′; sequence 2 forward, 5′-CCACCAGUUUGAAUGGUCAAUGCGA-3′ and reverse, 5′-UCGCAUUGACCAUUCAAACUGGUGG-3′; sequence 3 forward, 5′-CCGCCAAAUUCAACGGGCUCUUGUG-3′) and reverse, 5′-CACAAGAGCCCGUUGAAUUUGGCGG-3′). Control duplexes using Invitrogen stealth RNAi negative control duplexes (High GC Duplex, cat no. 12935-400) were utilized. The transfection procedure was performed following manufacturer's instructions. Cells were harvested at the same time for investigation after 24 to 48 h of growth.

Wound healing assay

Wound-healing assays were performed to assess the effect of migration. Gastric cancer cells were seeded into 6-well plates and treated with mitomycin C (Santa Cruz Biotechnology, Dallas, Texas, USA) to inhibit cell proliferation. The cell monolayer was disrupted with a pipette tip. DMEM medium was used to wash away floating cells. Photographs were captured using an inverted microscope (same magnification) at the same time 48 h after the scratch. Six fields for each point were recoded. Relative wound size was calculated to assess migration activity.

Cell invasion assay

For invasion assay, a Transwell assay was performed (8 µm; Corning Inc., New York, NY, USA). The membranes were coated with 200 µl Matrigel at 200 µg/ml. The upper chamber was seeded with cells in serum-free DMEM medium, and DMEM with 10% FBS was added in the lower chamber. After incubation for 24 h, cells were removed at the same time from the upper surface of the filter by scraping gently with a swab. Cells that invaded the bottom of membrane were fixed and stained. The numbers of invaded cells were calculated.

Western blot analysis

Cells were lysed in buffer [1% nonidet P-40, 100 mg/l phenylmethylsulfonyl fluoride, 50 mmol/l Tris-Cl (pH 8.0), 0.02% sodium azide, and 1 mg/l aprotinin]. After centrifugation for 20 min, the supernatant was collected, and the BCA protein assay kit (Beyotime Institute of Biotechnology, Shanghai, China) was used to measure protein concentrations following manufacturer's instructions. Equivalent amounts of protein were separated by 10% SDS-polyacrylamide gel electrophoresis. Then, proteins were transferred to a polyvinylidene fluoride (PVDF) membrane (Amersham Biosciences, Piscataway, NJ, USA) and blocked for 2 h at 37°C. The membranes were then incubated overnight at 4°C with primary antibodies. Immunocomplexes were incubated at room temperature with anti-mouse or anti-rabbit IgG for 1 h (diluted at 1:1,000). The results were visualized using an ECL kit (Amersham Biosciences). All antibodies and reagents were used based on manufacturer's instruction.

Statistical analysis

Data were analysed and presented as the means ± standard deviation (SD) of at least 3 independent experiments using one-way analysis of variance (one-way ANOVA). All analyses were performed using SPSS 13.0 software (SPSS Inc., Chicago, IL, USA). A p-value <0.05 was considered to indicate a significant difference.

Results

Notch1 is silenced by siRNA

The gastric cancer cell lines SGC7901 and MKN74 were transiently transfected with Notch1 siRNA and mock siRNA. We designed candidate Notch1 siRNA and negative control sequences (mock). Real-time PCR and western blot analysis were performed to assess the efficiency of Notch1 siRNA. As illustrated in Fig. 1, Notch1 was expressed in both SGC7901 and MKN74 cell lines, and the candidate sequence inhibited both Notch1 mRNA and protein expression compared to the control and mock treatments. Collectively, the expression of Notch1 protein was markedly decreased in the cells transfected with Notch1 siRNA compared with control (no siRNA) and mock (negative control siRNA) treatments in both cell lines (n=3, P<0.05).

The metastasis and invasion of gastric cancer cell lines were inhibited after downregulation of Notch1 expression

To determine whether the migratory abilities of SGC7901 and MKN74 cell lines were affected by Notch1 depletion, we performed wound-healing assays as presented in Fig. 2. The metastasis of SGC7901 was significantly suppressed after downregulation of Notch1 (Fig. 2A). The relative wound size of the Notch1 siRNA group (0.79±0.06 mm) was larger than the control (0.23±0.14 mm) and mock groups (0.19±0.12 mm) (n=6, P<0.05). A similar result was observed for MKN74 (Fig. 2B). The relative wound size of the Notch1 siRNA group (0.66±0.06 mm) was larger than the control (0.32±0.04 mm) and mock groups (0.33±0.05 mm) (n=6, P<0.05). These data demonstrated that Notch1 depletion inhibits the migration of SGC7901 and MKN74 cells. The results of Transwell invasion assays were consistent with the wound-healing assay results (Fig. 3). The number of invaded SGC7901 cells transfected with Notch1 siRNA (62±4.1) was significantly reduced compared with the control (148.5±11.4) and mock groups (147.3±8.0) (n=6 P<0.05) (Fig. 3A). The number of invaded MKN74 cells transfected with Notch1 siRNA (51.3±6.0) was also significantly reduced compared with the control (142.3±10.0) and mock groups (152.7±10.4) (n=6 P<0.05) (Fig. 3B). Taken together, our data indicate the role of Notch1 regarding invasion and metastasis in SGC7901 and MKN74 gastric cancer cells.

Inhibition of Notch1 alters expression of PTEN, pPTEN, pAkt and pFAK

As shown in Fig. 4, both mRNA and protein expression of PTEN was upregulated in the Notch1 siRNA group compared with the control and mock groups, whereas phospho-PTEN expression was downregulated after inhibition of Notch1 (n=3, P<0.05) in SGC7901 and MKN74 gastric cancer cell lines. The mRNA and protein expression of total Akt and FAK showed no significant changes. These results demonstrate that PTEN function is activated by the depletion of Notch1. Decreased expression of phospho-Akt and phospho-FAK but not total expression of Akt and FAK was also observed following reactivation of PTEN in both cell lines as shown in Fig. 4 (n=3, P<0.05).

Discussion

Invasion and metastasis are both vital causes of mortality in gastric cancer patients. Therefore, developing new treatments targeting invasion and metastasis are of great importance. Inhibition of cell signalling pathways shows great promise. As an upstream signalling pathway, the importance of Notch activation has been reported in numerous cancers (21,22). Increasing evidence has indicated that Notch1 is aberrantly activated and highly expressed in gastric cancer tissue (22,28). Notch1 plays an important tumor progression role in gastric cancer (22,29). Several studies have found that Notch1 promotes invasion and metastasis in cancer cells (3034). To examine whether Notch1 affects invasion and metastasis in gastric cancer cells, we used siRNA to inhibit Notch1 signalling in SGC7901 and MKN74. Notch signalling can be inhibited by prevention of ligand binding using gamma-secretase inhibitors (GSIs) or transcriptional activity inhibition. The methods we used to inhibit Notch1 in this study are widely effective, and we investigated the effect of inhibition using real-time PCR and western blot analysis. We also employed wound-healing and Transwell assays to assess the effect of cell invasion and metastasis after Notch1 downregulation. The data indicate that siRNA downregulates the expression of Notch1 mRNA and protein following the suppression of cell invasion and metastasis of SCG7901 and MKN74 gastric cancer cells. This result indicates the role of Notch1 in gastric cancer cell lines SCG7901 and MKN74 regarding invasion and metastasis in vitro and also suggests that Notch1 could be a potential therapeutic target in gastric cancer treatment.

To explore the mechanism by which Notch1 affects invasion and metastasis in SGC7901 and MKN74 cells, we focused on expression of PTEN and phospho-PTEN (Ser380/Thr382/383). PTEN acts as a tumor suppressor by functioning as a dual-specificity protein and phospholipid phosphatase (35). Its function depends on its protein structure, which has five distinct domains: N-term, Phosphatase domain, C2 domain, C-tail and PDZ. N-term contains the PIP binding domain. The phosphatase domain is responsible for its enzymatic and phosphatase activity. The C2 domain is responsible for its cellular location and protein-protein interactions. The C-tail domain is less defined but may be critical for the stability of PTEN and the C-terminal PDZ domain (36). PTEN is involved in numerous biological processes, and its regulation is very complex. One of its important regulations is posttranslational. The C-terminal tail of PTEN can be phosphorylated at Ser380, Thy382 and Thy383. The result of this regulation is inhibition of PTEN's critical phosphatase activity, thus leading to cell growth promotion (37). Phospho-PTEN (Ser380/Thr382/383) protein expression may inhibit PTEN activation.

In this study, increased total PTEN mRNA and protein expression and decreased phospho-PTEN expression was observed following Notch1 depletion. Notch can either be a tumor promoter or tumor suppressor via differential regulation of PTEN protein expression in different situations (38,39). In this situation, Notch1 acts as a tumor promoter that negatively correlates with PTEN expression and positively correlates with phospho-PTEN (Ser380/Thr382/383) protein expression.

We hypothesize that Notch1 negatively regulates PTEN activation not only by suppressing total PTEN expression but also by phosphorylating the PTEN C-terminal tail at Ser380, Thy382 and Thy383, thus causing inhibition of PTEN's phosphatase activity in gastric cancer cell lines. This hypothesis is supported by Kim et al research; they determined that Notch signalling disables PTEN by phosphorylation and contributes to tumorigenesis (40). We will investigate relative mechanism of this regulation and focus on testing if PTEN activation is required by inhibition of migration in gastric cancer cells upon depletion of Notch1 in our further study.

Cancer cell invasion and metastasis involves many mechanisms. Activation of Akt and FAK signalling pathways through phosphorylation promotes invasion, metastasis and proliferation (4143). One of the classic PTEN functions involves dephosphorylating PIP3, thus antagonizing the (PI3K)/Akt signalling pathway (16). PTEN also downregulates the activity of FAK by dephosphorylation (18). In this study, decreased expression of phosphorylated Akt and FAK was observed after the inhibition of Notch1. We hypothesized that decreased expression of phosphorylated Akt and FAK directly leads to suppression of cell invasion and metastasis and correlates with re-activation of PTEN. However, further investigations are required.

Collectively, our results demonstrate that invasion and metastasis in SGC7901 and MKN74 gastric cancer cells are inhibited in vitro after downregulation of the Notch1 signalling pathway by siRNA. Depletion of Notch1 leads to increased PTEN and decreased phospho-PTEN (Ser380/Thr382/383) protein expression in gastric cancer cells. Re-activation of PTEN by inhibition of Notch1 leads to decreased expression of phosphorylated Akt and FAK. The Notch1-PTEN-Akt&FAK signalling axis may serve as a further treatment of gastric cancer targeting invasion and metastasis.

Acknowledgements

We thank the Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education (Harbin Medical University). Our entire experiment was conducted in this laboratory, and we were given useful advice by lab working staff. We also thank The 2nd Affiliated Hospital of Harbin Medical University and Harbin Medical University Cancer Hospital for kindly providing SGC7901 and MKN74 gastric cell lines.

References

1 

Karimi P, Islami F, Anandasabapathy S, Freedman ND and Kamangar F: Gastric cancer: Descriptive epidemiology, risk factors, screening and prevention. Cancer Epidemiol Biomarkers Prev. 23:700–713. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Brenner H, Rothenbacher D and Arndt V: Epidemiology of stomach cancer. Methods Mol Biol. 472:467–477. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Yang L: Incidence and mortality of gastric cancer in China. World J Gastroenterol. 12:17–20. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Zhang XB, Song L, Wen HJ, Bai XX, Li ZJ and Ma LJ: Upregulation of microRNA-31 targeting integrin α5 suppresses tumor cell invasion and metastasis by indirectly regulating PI3K/AKT pathway in human gastric cancer SGC7901 cells. Tumor Biol. 37:8317–8325. 2016. View Article : Google Scholar

8 

Liu JJ, Liu JY, Chen J, Wu YX, Yan P, Ji CD, Wang YX, Xiang DF, Zhang X, Zhang P, et al: Scinderin promotes the invasion and metastasis of gastric cancer cells and predicts the outcome of patients. Cancer Lett. 376:110–117. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Deng QJ, Xie LQ and Li H: Overexpressed MALAT1 promotes invasion and metastasis of gastric cancer cells via increasing EGFL7 expression. Life Sci. 157:38–44. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Tan C, Qiao F, Wei P, Chi Y, Wang W, Ni S, Wang Q, Chen T, Sheng W, Du X and Wang L: DIXDC1 activates the Wnt signaling pathway and promotes gastric cancer cell invasion and metastasis. Mol Carcinog. 55:397–408. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Li J, Deng Z, Wang Z, Wang D, Zhang L, Su Q, Lai Y, Li B, Luo Z, Chen X, et al: Zipper-interacting protein kinase promotes epithelial-mesenchymal transition, invasion and metastasis through AKT and NF-κΒ signaling and is associated with metastasis and poor prognosis in gastric cancer patients. Oncotarget. 6:8323–8338. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Bertrand FE, McCubrey JA, Angus CW, Nutter JM and Sigounas G: NOTCH and PTEN in prostate cancer. Adv Biol Regul. 56:51–65. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Steck PA, Pershouse MA, Jasser SA, Yung WK, Lin H, Ligon AH, Langford LA, Baumgard ML, Hattier T, Davis T, et al: Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet. 15:356–362. 1997. View Article : Google Scholar : PubMed/NCBI

14 

Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C, Rodgers L, McCombie R, et al: PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 275:1943–1947. 1997. View Article : Google Scholar : PubMed/NCBI

15 

Li DM and Sun H: TEP1, encoded by a candidate tumor suppressor locus, is a novel protein tyrosine phosphatase regulated by transforming growth factor beta. Cancer Res. 57:2124–2129. 1997.PubMed/NCBI

16 

Milella M, Falcone I, Conciatori F, Incani U Cesta, Del Curatolo A, Inzerilli N, Nuzzo CM, Vaccaro V, Vari S, Cognetti F and Ciuffreda L: PTEN: Multiple functions in human malignant tumors. Front Oncol. 5:242015. View Article : Google Scholar : PubMed/NCBI

17 

Tamura M, Gu J, Takino T and Yamada KM: Tumor suppressor PTEN inhibition of cell invasion, migration, and growth: Differential involvement of focal adhesion kinase and p130Cas. Cancer Res. 59:442–449. 1999.PubMed/NCBI

18 

Tamura M, Gu J, Matsumoto K, Aota S, Parsons R and Yamada KM: Inhibition of cell migration, spreading, and focal adhesions by tumor suppressor PTEN. Science. 280:1614–1617. 1998. View Article : Google Scholar : PubMed/NCBI

19 

Wang S, Tie J, Wang R, Hu F, Gao L, Wang W, Wang L, Li Z, Hu S, Tang S, et al: SOX2, a predictor of survival in gastric cancer, inhibits cell proliferation and metastasis by regulating PTEN. Cancer Lett. 358:210–219. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Wang H, Wu Q, Liu Z, Luo X, Fan Y, Liu Y, Zhang Y, Hua S, Fu Q, Zhao M, et al: Downregulation of FAP suppresses cell proliferation and metastasis through PTEN/PI3K/AKT and Ras-ERK signaling in oral squamous cell carcinoma. Cell Death Dis. 5:e11552014. View Article : Google Scholar : PubMed/NCBI

21 

Lobry C, Oh P and Aifantis I: Oncogenic and tumor suppressor functions of Notch in cancer: It's NOTCH what you think. J Exp Med. 208:1931–1935. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Du X, Cheng Z, Wang YH, Guo ZH, Zhang SQ, Hu JK and Zhou ZG: Role of Notch signaling pathway in gastric cancer: A meta-analysis of the literature. World J Gastroenterol. 20:9191–9199. 2014.PubMed/NCBI

23 

Baron M, Aslam H, Flasza M, Fostier M, Higgs JE, Mazaleyrat SL and Wilkin MB: Multiple levels of Notch signal regulation (Review). Mol Membr Biol. 19:27–38. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Grishina IB: Mini-review: Does Notch promote or suppress cancer? New findings and old controversies. Am J Clin Exp Urol. 3:24–27. 2015.PubMed/NCBI

25 

Yap LF, Lee D, Khairuddin A, Pairan MF, Puspita B, Siar CH and Paterson IC: The opposing roles of NOTCH signalling in head and neck cancer: A mini review. Oral Dis. 21:850–857. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Kwon OJ, Zhang L, Wang J, Su Q, Feng Q, Zhang XH, Mani SA, Paulter R, Creighton CJ, Ittmann MM and Xin L: Notch promotes tumor metastasis in a prostate-specific Pten-null mouse model. J Clin Invest. 126:2626–2641. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Hu YJ, Li HY, Qiu KJ, Li DC, Zhou JH, Hu YH and Zhang FM: Downregulation of Notch1 inhibits the invasion of human hepatocellular carcinoma HepG2 and MHCC97H cells through the regulation of PTEN and FAK. Int J Mol Med. 34:1081–1086. 2014.PubMed/NCBI

28 

Wu X, Liu W, Tang D, Xiao H, Wu Z, Chen C, Yao X, Liu F and Li G: Prognostic values of four Notch receptor mRNA expression in gastric cancer. Sci Rep. 6:280442016. View Article : Google Scholar : PubMed/NCBI

29 

Yeh TS, Wu CW, Hsu KW, Liao WJ, Yang MC, Li AF, Wang AM, Kuo ML and Chi CW: The activated Notch1 signal pathway is associated with gastric cancer progression through cyclooxygenase-2. Cancer Res. 69:5039–5048. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Jia M, Jiang L, Wang YD, Huang JZ, Yu M and Xue HZ: lincRNA-p21 inhibits invasion and metastasis of hepatocellular carcinoma through Notch signaling-induced epithelial-mesenchymal transition. Hepatol Res. 46:1137–1144. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Zhao ZL, Ma SR, Wang WM, Huang CF, Yu GT, Wu TF, Bu LL, Wang YF, Zhao YF, Zhang WF and Sun ZJ: Notch signaling induces epithelial-mesenchymal transition to promote invasion and metastasis in adenoid cystic carcinoma. Am J Transl Res. 7:162–174. 2015.PubMed/NCBI

32 

Sonoshita M, Itatani Y, Kakizaki F, Sakimura K, Terashima T, Katsuyama Y, Sakai Y and Taketo MM: Promotion of colorectal cancer invasion and metastasis through activation of NOTCH-DAB1-ABL-RHOGEF protein TRIO. Cancer Discov. 5:198–211. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Zhang P, Yang Y, Zweidler-McKay P and Hughes DP: Retraction: Critical role of notch signaling in osteosarcoma invasion and metastasis. Clin Cancer Res. 14:2962–2969. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Zhang P, Yang Y, Zweidler-McKay PA and Hughes DP: Critical role of notch signaling in osteosarcoma invasion and metastasis. Clin Cancer Res. 14:2962–2969. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Xu WT, Yang Z and Lu NH: Roles of PTEN (phosphatase and tensin homolog) in gastric cancer development and progression. Asian Pac J Cancer Prev. 15:17–24. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Jerde TJ: Phosphatase and tensin homologue: Novel regulation by developmental signaling. J Signal Transduct. 2015:2825672015. View Article : Google Scholar : PubMed/NCBI

37 

Odriozola L, Singh G, Hoang T and Chan AM: Regulation of PTEN activity by its carboxyl-terminal autoinhibitory domain. J Biol Chem. 282:23306–23315. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Whelan JT, Kellogg A, Shewchuk BM, Hewan-Lowe K and Bertrand FE: Notch-1 signaling is lost in prostate adenocarcinoma and promotes PTEN gene expression. J Cell Biochem. 107:992–1001. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Chappell WH, Green TD, Spengeman JD, McCubrey JA, Akula SM and Bertrand FE: Increased protein expression of the PTEN tumor suppressor in the presence of constitutively active Notch-1. Cell Cycle. 4:1389–1395. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Kim SJ, Lee HW, Baek JH, Cho YH, Kang HG, Jeong JS, Song J, Park HS and Chun KH: Activation of nuclear PTEN by inhibition of Notch signaling induces G2/M cell cycle arrest in gastric cancer. Oncogene. 35:251–260. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Zhao HF, Wang J, Jiang HR, Chen ZP and To SS: PI3K p110β isoform synergizes with JNK in the regulation of glioblastoma cell proliferation and migration through Akt and FAK inhibition. J Exp Clin Cancer Res. 35:782016. View Article : Google Scholar : PubMed/NCBI

42 

Gao H, Zhong F, Xie J, Peng J and Han Z: PTTG promotes invasion in human breast cancer cell line by upregulating EMMPRIN via FAK/Akt/mTOR signaling. Am J Cancer Res. 6:425–439. 2016.PubMed/NCBI

43 

Egawa H, Jingushi K, Hirono T, Ueda Y, Kitae K, Nakata W, Fujita K, Uemura M, Nonomura N and Tsujikawa K: The miR-130 family promotes cell migration and invasion in bladder cancer through FAK and Akt phosphorylation by regulating PTEN. Sci Rep. 6:205742016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 16 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang XS, Hu YH, Gao HY, Lan XW and Xue YW: Downregulation of Notch1 inhibits the invasion and metastasis of human gastric cancer cells SGC7901 and MKN74 in vitro through PTEN activation and dephosphorylation of Akt and FAK. Mol Med Rep 16: 2318-2324, 2017
APA
Zhang, X., Hu, Y., Gao, H., Lan, X., & Xue, Y. (2017). Downregulation of Notch1 inhibits the invasion and metastasis of human gastric cancer cells SGC7901 and MKN74 in vitro through PTEN activation and dephosphorylation of Akt and FAK. Molecular Medicine Reports, 16, 2318-2324. https://doi.org/10.3892/mmr.2017.6791
MLA
Zhang, X., Hu, Y., Gao, H., Lan, X., Xue, Y."Downregulation of Notch1 inhibits the invasion and metastasis of human gastric cancer cells SGC7901 and MKN74 in vitro through PTEN activation and dephosphorylation of Akt and FAK". Molecular Medicine Reports 16.2 (2017): 2318-2324.
Chicago
Zhang, X., Hu, Y., Gao, H., Lan, X., Xue, Y."Downregulation of Notch1 inhibits the invasion and metastasis of human gastric cancer cells SGC7901 and MKN74 in vitro through PTEN activation and dephosphorylation of Akt and FAK". Molecular Medicine Reports 16, no. 2 (2017): 2318-2324. https://doi.org/10.3892/mmr.2017.6791