IL-8 suppresses E-cadherin expression in nasopharyngeal carcinoma cells by enhancing E-cadherin promoter DNA methylation

  • Authors:
    • Rui-Li Zhang
    • Li-Xia Peng
    • Jun-Ping Yang
    • Li-Sheng Zheng
    • Ping Xie
    • Meng-Yao Wang
    • Bi-Jun Huang
    • Hua-Rong Zhao
    • Yong-Xing Bao
    • Chao-Nan Qian
  • View Affiliations

  • Published online on: November 2, 2015     https://doi.org/10.3892/ijo.2015.3226
  • Pages: 207-214
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Nasopharyngeal carcinoma (NPC) has the highest metastasis potential among head and neck cancers. Distant metastasis is the major cause of treatment failure. Recent studies from our laboratory have revealed that IL-8 promotes NPC metastasis via activation of AKT signaling and induction of epithelial-mesenchymal transition (EMT) in the cells. In the present study, we found that IL-8 treatment for NPC cells resulted in an accumulation of DNMT1 protein through activating AKT1 pathway and consequent DNMT1 protein stabilization. Then DNMT1 suppressed E-cadherin expression by increasing the methylation of its promoter region. LY-294002 blocked IL-8-induced p-AKT1 activation resulting in reduction of DNMT1 and increase of E-cadherin expression, whereas forced demethylation using 5-aza-2'-deoxycytidine restored E-cadherin expression. In conclusion, our study, for the first time, shows that the IL-8/AKT1 signaling pathway stabilizes DNMT1 protein, consequently enhancing hypermethylation of E-cadherin promoter regions and downregulating E-cadherin protein level in NPC cells. Upon blockage of the IL-8/AKT pathway and inhibition of DNMT1, E-cadherin expression can be reversed. These data suggest that targeting the IL-8/AKT1 signaling pathway and DNMT1 may provide a potential therapeutic approach for blocking NPC metastasis.

Introduction

Nasopharyngeal carcinoma (NPC) has a high incidence rate in southern China and Southeast Asia, and its metastasis rate is also the highest among head and neck cancers (13). The close relationship of Epstein-Barr virus infection suggests NPC as an inflammation-associated cancer (4,5).

Interleukin 8 (IL-8; alternatively known as CXCL8) is a proinflammatory cysteine-X-cysteine (CXC) chemokine, plays multiple roles by mediating the activation and chemotaxis of various immune cell types, to promote immune infiltration and angiogenesis, which in turn establishes a venue for cancer cell local invasion, migration, and metastasis. Studies have shown that IL-8 promotes tumor growth and metastasis in melanoma (69), bladder cancer (10), and ovarian cancer (11). We have also previously demonstrated that the overexpression of IL-8 in nasopharyngeal carcinoma cell line S26 cells and HONE-1 cells activated AKT1 signaling and induced EMT (12). In recent years, it has also been demonstrated that a link exists between IL-8 and tumor EMT, which involve decreased expression of epithelial markers such as E-cadherin in lung cancer (13), hepatocellular carcinoma (14,15) and thyroid cancer (16).

E-cadherin is a key mediator of cell-cell adhesion in epithelial tissues, and loss of E-cadherin can promote invasive and metastatic behavior in many epithelial tumors (17). In head and neck cancers, loss of cell-cell adhesion resulting in stromal and vascular invasion as a consequence of E-cadherin dysregulation is well documented (18,19).

It has been suggested that DNA methylation plays a major role in enhancing transcriptional silence, especially in tumor suppressor genes (20). E-cadherin also could be suppressed by DNA hypermethylation and has a close relationship with tumor prognosis in head and neck squamous cell carcinoma (21), breast cancer (22), lung cancer (23), bladder cancer (24).

DNA methyltransferases (DNMTs) are responsible for the transfer of a methyl group from the universal methyl donor, S-adenosyl-L-methionine, to the 5-position of the cytosine residue in DNA. There are four members of the DNMT family: DNMT1, DNMT3A, DNMT3B and DNMT3L. DNMT3A and DNMT3B encode de novo methyltransferases, while DNMT1 encodes a maintenance methyltransferases, which are essential for mammalian development and reported to be associated with human tumorigenesis (25).

DNMT1 stability is regulated via various post-translational modifications, previous studies showed that AKT1 can stabilizes DNMT1 and affects genome methylation (26,27). IL-8 can activate AKT1 pathway to promote tumor invasion in breast cancer (28), kidney cancer (29), and breast cancer (30). Our previous study also showed that in NPC, the overexpression of IL-8 can induce EMT through activating the AKT signaling pathway (12). However, to the best of our knowledge, there are no studies regarding the relationship between IL-8 and DNMT1 expression, and though our previous study showed IL-8 can reduce E-cadherin expression in NPC cell lines (12), the underlying molecular mechanisms remain unclear. In this study, we explore the possibility that in NPC cell lines, IL-8 can induce DNMT1 expression through AKT1 signaling then mediates silencing of E-cadherin expression which play an important role in EMT.

Materials and methods

Cell lines and cell culture

The human low-metastasis, low endogenous IL-8 secreating level NPC cell lines CNE-2 and the CNE-2 subclones S22 and S26 have previously been established and reported (12,31), the cells were cultured in Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum purchased from Gibco/BRL (Grand Island, NY, USA), 100 U/ml penicillin G, 100 U of streptomycin (all from Invitrogen, Carlsbad, CA, USA). Cells were incubated at 37°C in a humidified atmosphere containing 5% CO2. Based on our previous study (12), recombinant human IL-8 (PeproTech) at a concentration of 8 ng/ml was added into cell culture medium in experimental groups. For vehicle controls, the cells were treated with equivalent amounts of BSA.

Immunoblotting

Immunoblotting was performed as described previously (12). The sources of the primary antibodies (and their concentrations) were as follows: anti-E-cadherin (1:1,000), anti-AKT (1:1,000), anti-phospho-AKT (Ser473) (1:1,000), and anti-β-actin (1:1,000) these antibodies were purchased from Cell Signaling Technology (Danvers, MA, USA); anti-DNMT1 (1:1,000) was purchased from Santa Cruz Biotechnology (Santa Cruz). Anti-rabbit peroxidase-conjugated secondary antibodies were purchased from Promega. For western blot analysis of AKT1 signaling inhibition studies, LY-294002 (the AKT1 inhibitor) (Cell Signaling Technology) (20 μM) was used one hour prior to IL-8 stimulation in an attempt to inhibit the conventional AKT1 pathway, for western blot analysis of DNMT1 inhibition studies, 5-aza-2′-deoxycytidine (the inhibitor of DNA methylation) (Sigma) (5 μM) was used in an attempt to inhibit DNMT1 function. Cells were pretreated with 5-aza-2′-deoxycytidine for 48 h then were treated with IL-8 for 24 h. Experiments were repeated in triplicate.

Quantitative real-time polymerase chain reaction

Total cellular RNA was extracted using the High Pure RNA kit (Roche Applied Science, Penzberg, Germany). For RT-PCR, the total RNA was quantified spectrophotometrically, and equal amounts (1 μg) were transcribed into cDNA according to the manufacturer's protocol (Roche Applied Science). The sequences of the PCR primers used for amplifications of β-actin, IL-8, DNMT1, E-cadherin were as follows: β-actin forward: 5′-CACGATGGAGGGGCCGGACTCATC-3′; β-actin reverse, 5′-TAAAGACCTCTATGCCAACACAGT-3′. IL-8 forward, 5′-CTCCAAACCTTTCCACCCC-3′; IL-8 reverse, 5′-GATTCTTGGATACCACAGAGAATG-3′. DNMT1 forward, 5′-AGCCAAATCGGATGAGTCCATC-3′; DNMT1 reverse, 5′-CCTCCTTCAGTTTCTGTTTGGG-3′. E-cadherin forward, 5′-AACAGGATGGCTGAAGGTGA-3′; E-cadherin reverse, 5′-CCTTCCATGACAGACCCCTT-3′. Fast SYBR Green Master Mix was used to determine the threshold cycle (Ct) value of each sample in the CFX96 real-time PCR detection system (Bio-Rad, CA, USA). β-actin served as the normalization gene in these studies. The relative expression levels of the target genes were given by 2ΔCt (Ct of β-actin minus the Ct of the target gene). PCR amplifications of β-actin, IL-8, E-cadherin, DNMT1 were performed under the following conditions: denaturation at 95°C for 30 sec, annealing at 58°C for 30 sec, and extension at 72°C for 30 sec; reactions were carried out for 30 cycles. The experiments were performed in triplicate.

Transient transfection of IL-8

For IL-8 overexpression studies, the IL-8 overexpressing vector (cat no. CH832510) and control pENTER vector were purchased from ViGene Biosciences Inc. (Rockville, MD, USA). These plasmids were transfected into S22, S26, CNE2 cells with X-tremeGENE HP transfection reagent according to the manufacturer's instructions (Roche Life Science). The cells were harvest 48 h after transfection for immunoblot and real time-PCR analyses.

ELISA

Twenty-four hours after IL-8 overexpressing plasmids or control pENTER vector were transfected into S22, S26, CNE2 cells with X-tremeGENE HP transfection reagent, 2×106 of the cells were plated into 100-mm culture plates and incubated for another 24 h in the regular medium. The medium was then replaced with a serum-free medium (10 ml) and the cells were incubated for an additional 12 h. Conditioned medium was collected and subjected to centrifugation, followed by filtration through a 0.45 μm membrane filter to remove the debris. Secreted human IL-8 concentration in the conditioned medium was then measured using a sandwich enzyme-linked immunosorbent assay (ELISA) kit (R&D, MN, USA) following the manufacturer's instructions. The experiments were performed in triplicate.

BSP (bisulfite sequencing PCR)

Bisulfite sequencing PCR was used to examine the methylation status of E-cadherin gene promoter. Genomic DNA from cultured cells were extracted according to the manufacturer's protocol (Qiagen 51304), followed by bisulfite conversion using EpiTect Bisulfite kit (Qiagen 59104). DNA amplification was performed via PCR using primers as follows: E-cadherin-BSP-F: 5′-TGTAGGT TTTATAATTTATTTAGAT-3′; E-cadherin-BSP-R: 5′-CTCA CAAATACTTTACAATT-3′. PCR products were cloned to pTopo TA vector (Invitrogen), and then transfected to Top10 cells for sequencing, at least ten clones from each sample were selected.

Statistics

Student's t-test was used to compare two independent groups of data. A P-value <0.05 was considered statistically significant.

Results

IL-8 treatment induces DNMT1 expression in nasopharyngeal cancer cells

Our previous study showed that IL-8 is lowly expressed in low-metastasis S26 cells (12). In the present study, we investigated whether IL-8 could enhance the expression of DNMT1 by transient transfection of IL-8 plasmid. After transient transfection of IL-8 plasmid in S22, S26, and CNE-2 cells, real-time PCR analysis showed that IL-8 mRNA levels were increased greatly 48 h later (Fig. 1A), and ELISA analysis showed high levels of IL-8 were secreted into the culture medium by the cells (Fig. 1B). Then immunoblotting of the nuclear extracts showed an increase in DNMT1 protein expression 48 h after transient transfections (Fig. 1C). Next, we used exogenous recombinant human IL-8 to verify this finding. Consistently, DNMT1 protein level was increased in the S22, S26, CNE-2 cells following IL-8 treatment with 8 ng/ml for 24 h (Fig. 1D). We next analyzed DNMT1 mRNA expression using real-time PCR, and found that DNMT1 mRNA levels were not significantly altered in these cells (Fig. 1E), suggesting that IL-8-induced accumulation of DNMT1 protein was not due to mRNA overexpression.

IL-8 enhances DNMT1 stabilization through AKT1 signaling

DNMT1 protein stability can be regulated via various post-translational modifications. It has been reported that AKT1 activity can stabilize DNMT1 protein (26,27). Our previous study showed that IL-8 can stimulate AKT1 pathway in NPC cells (12), therefore, we wanted to know whether the IL-8-induced accumulation of DNMT1 protein is partly due to AKT1 signaling. After transient transfection of IL-8, immunoblotting of nuclear extracts showed an increase in p-AKT1 protein expression in S22, S26, and CNE-2 cells (Fig. 2A). Then S22, S26, CNE-2 cells were treated with IL-8 at 8 ng/ml, and total AKT1 and phosphorylated AKT1 levels were tested by immunoblotting. The results showed that treatment of cells with IL-8 lead to activation of the AKT1 pathway as expected. To confirm that the increase of DNMT1 is AKT1-dependent, we treated cultured S22, S26, CNE-2 cells with the AKT1 inhibitor LY294002 and then measured DNMT1 protein levels. LY294002 blocked pAKT1 activation triggered by IL-8 and resulted in reduction of total DNMT1 (Fig. 2B). To confirm DNMT1 protein stabilization, we treated S26 cells with the protein synthesis inhibitor cycloheximide to block the synthesis of DNMT1. Immunoblot analysis showed that DNMT1 levels gradually decreased in 10 h following the addition of cycloheximide to culture media without IL-8. However, the degradation of DNMT1 was slowed down by the addition of IL-8 to the medium, indicating that IL-8 stabilizes DNMT1 protein, an effect persisting for ≥12 h (Fig. 2C). Therefore, we conclude that IL-8 upregulates DNMT1 protein level through activating AKT1 pathway and enhancing DNMT1 stabilization.

IL-8 treatment of S26 cells increases E-cadherin level via DNMT1

Our previous study shows that overexpression of IL-8 in S26 cells can induce EMT with downregulation of E-cadherin (12). In the present study, we continued to explore whether this downregulation of E-cadherin level is partly due to upregulation of DNMT1. Immunoblot analysis showed that IL-8 inhibited the expression of E-cadherin in S26 cell lines and also decreased the transcription of E-cadherin (Fig. 3A and B). As inhibiting AKT1 reduced the overall DNMT1 protein levels, we expected that treating the cells with LY294002 would result in upregulated E-cadherin expression. Then, S26 cells were treated with IL-8 for 24 h, while in the presence of LY-294002, an inhibitor of the AKT pathway, which was added one hour before the addition of IL-8, immunoblot analysis showed that LY-294002 blocked IL-8-induced pAKT1 activation and resulted in reduction of DNMT1 as well as an increase of E-cadherin protein level. Many studies have demonstrated that promoter methylation of E-cadherin is an important mechanism contributing to its downregulation (33). We speculate that methylation of the E-cadherin promoter may play an important role in IL-8 mediated decrease of this gene. In addition, while in the presence of 5-aza-2′-deoxycytidine, an inhibitor of DNA methylation, which was added 48 h before IL-8, and IL-8-induced downregulation of E-cadherin was reversed (Fig. 3C). The RNA level of E-cadherin also showed consistent changes (Fig. 3D), suggesting that the effect of IL-8 on E-cadherin expression is transcriptionally regulated via DNMT1.

IL-8 downregulates E-cadherin expression by promoting its promoter methylation via DNMT1

Next, in order to verify that the downregulation of E-cadherin was partly due to its promoter methylation, the bisulfite sequencing PCR of E-cadherin promoter analysis was performed, the methylated cytosine residues (ranging from −280 to +40 of E-cadherin promoter) in each clone were represented by a solid spot as depicted (Fig. 4). In total, 17 CpG methylation sites were detected. Bisulfite sequencing PCR assay of the E-cadherin promoter showed that the promoter CpG island methylation status of E-cadherin in IL-8 treated cells was markedly higher than control cells (Fig. 4). In addition, when LY-294002 or 5-aza-2′-deoxycytidine was added for 48 h together with IL-8 administration, the methylation status of E-cadherin reversed to the control level. These data demonstrated that IL-8 could induce hypermethylation on the specific CpG sites of E-cadherin promoter.

Discussion

The influence of inflammation on tumorigenesis has been intensively investigated for centuries (34). In addition, the expression of IL-8 in the tumor microenvironment is reported to be associated with tumor progression and patient survival (35). We have previously reported that IL-8 promotes metastasis of NPC cells in autocrine and paracrine manner, involving activation of AKT signaling and inducing EMT in NPC cells (12). In the present study, we further demonstrated for the first time that IL-8 inhibited the expression of E-cadherin by inducing its promoter DNA hypermethylation via upregulating DNMT1 protein level. Blockage of the IL-8/AKT pathway and inhibition of DNMT1 reversed the expression of E-cadherin. These data demonstrate a link between inflammation and NPC progression involving epigenetic regulation of E-cadherin.

Dysregulation of DNMT1 activity causes many human diseases, including cancer (36). Post-translational modifications of DNMT1 play a crucial role in how and when it is activated. Other studies have reported that AKT activity can inhibit DNMT1 degradation in multiple cell lines (26,27). IL-8 signals through CXCR1 and CXCR2, and phosphatidylinositol-3 (PI3) is a component in CXCR1/2-signaling. The enzyme PI3-kinase (PI3K) is a principal effector of CXCL8-mediated chemotaxis in neutrophils, and its triggering phosphorylation results in the activation and increased expression of AKT (28,32,37,38). However, to our knowledge, our study is the first one to clarify the relationship between IL-8 and DNMT1. Our findings showed that IL-8 upregulated DNMT1 though AKT1 pathway by increasing its stability in NPC cells. This increment of DNMT1 by IL-8 stimulation can be eliminated by blocking AKT signaling, confirming that AKT is a key signaling pathway by which IL-8 regulates DNMT1. Upon DNMT1 activation, both transcriptional and translational levels of E-cadherin are reduced. We therefore confirmed E-cadherin as one of the target genes of DNMT1 is regulated by IL-8/AKT signaling. The important roles of E-cadherin in NPC progression relies on its functions as an adhesion molecule regulating cell-cell contact for tissue morphogenesis, cellular polarity and tumor invasiveness (17). It has been reported that loss of membranous E-cadherin expression results in enhanced cell migration activity (39), which significantly correlates with tumor invasion, advanced disease stage, and tumor metastasis (40).

A model for this IL-8-p-AKT1-DNMT1 signaling is then proposed based on our findings (Fig. 5) our data clearly show that the IL-8-activated AKT signaling results in stabilization of DNMT1 and E-cadherin epigenetic regulation. This is the first report to reveal the IL-8-mediated E-cadherin silencing mechanism in NPC. These data suggest that targeting IL-8 signaling is a promising approach for prevention and treatment of NPC metastasis.

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (nos. 81472386, 81272340 and 81030043), the National High Technology Research and Development Program of China (863 Program, no. 2012AA02A501), and the Urumqi Key Laboratory of Infection and Cancer Project Foundation of China (no. WIT-2013-04).

Abbreviations:

NPC

nasopharyngeal carcinoma

DNMT1

DNA methyltransferase-1 interleukin

5-aza

5-aza-2′-deoxycytidine

EMT

epithelial-mesenchymal transition

References

1 

Wei KR, Zheng RS, Zhang SW, Liang ZH, Ou ZX and Chen WQ: Nasopharyngeal carcinoma incidence and mortality in China in 2010. Chin J Cancer. 33:381–387. 2014.PubMed/NCBI

2 

Adham M, Kurniawan AN, Muhtadi AI, Roezin A, Hermani B, Gondhowiardjo S, Tan IB and Middeldorp JM: Nasopharyngeal carcinoma in Indonesia: epidemiology, incidence, signs, and symptoms at presentation. Chin J Cancer. 31:185–196. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Tao CJ, Liu X, Tang LL, Mao YP, Chen L, Li WF, Yu XL, Liu LZ, Zhang R, Lin AH, et al: Long-term outcome and late toxicities of simultaneous integrated boost-intensity modulated radiotherapy in pediatric and adolescent nasopharyngeal carcinoma. Chin J Cancer. 32:525–532. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Liang FY, Sun W, Han P, Lu X, Lian YN and Huang XM: Detecting plasma Epstein-Barr virus DNA to diagnose postradiation nasopharyngeal skull base lesions in nasopharyngeal carcinoma patients: a prospective study. Chin J Cancer. 31:142–149. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Wang WY, Twu CW, Lin WY, Jiang RS, Liang KL, Chen KW, Wu CT, Shih YT and Lin JC: Plasma Epstein-Barr virus DNA screening followed by 18F-fluoro-2-deoxy-D-glucose positron emission tomography in detecting posttreatment failures of nasopharyngeal carcinoma. Cancer. 117:4452–4459. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Bar-Eli M: Role of interleukin-8 in tumor growth and metastasis of human melanoma. Pathobiology. 67:12–18. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Rofstad EK and Halsør EF: Vascular endothelial growth factor, interleukin 8, platelet-derived endothelial cell growth factor, and basic fibroblast growth factor promote angiogenesis and metastasis in human melanoma xenografts. Cancer Res. 60:4932–4938. 2000.PubMed/NCBI

8 

Singh RK, Gutman M, Reich R and Bar-Eli M: Ultraviolet B irradiation promotes tumorigenic and metastatic properties in primary cutaneous melanoma via induction of interleukin 8. Cancer Res. 55:3669–3674. 1995.PubMed/NCBI

9 

Luca M, Huang S, Gershenwald JE, Singh RK, Reich R and Bar-Eli M: Expression of interleukin-8 by human melanoma cells up-regulates MMP-2 activity and increases tumor growth and metastasis. Am J Pathol. 151:1105–1113. 1997.PubMed/NCBI

10 

Inoue K, Slaton JW, Kim SJ, Perrotte P, Eve BY, Bar-Eli M, Radinsky R and Dinney CP: Interleukin 8 expression regulates tumorigenicity and metastasis in human bladder cancer. Cancer Res. 60:2290–2299. 2000.PubMed/NCBI

11 

Shahzad MM, Arevalo JM, Armaiz-Pena GN, Lu C, Stone RL, Moreno-Smith M, Nishimura M, Lee JW, Jennings NB, Bottsford-Miller J, et al: Stress effects on FosB- and interleukin-8 (IL8)-driven ovarian cancer growth and metastasis. J Biol Chem. 285:35462–35470. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Li XJ, Peng LX, Shao JY, Lu WH, Zhang JX, Chen S, Chen ZY, Xiang YQ, Bao YN, Zheng FJ, et al: As an independent unfavorable prognostic factor, IL-8 promotes metastasis of nasopharyngeal carcinoma through induction of epithelial-mesenchymal transition and activation of AKT signaling. Carcinogenesis. 33:1302–1309. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Desai S, Laskar S and Pandey BN: Autocrine IL-8 and VEGF mediate epithelial-mesenchymal transition and invasiveness via p38/JNK-ATF-2 signalling in A549 lung cancer cells. Cell Signal. 25:1780–1791. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Fu XT, Dai Z, Song K, Zhang ZJ, Zhou ZJ, Zhou SL, Zhao YM, Xiao YS, Sun QM, Ding ZB, et al: Macrophage-secreted IL-8 induces epithelial-mesenchymal transition in hepatocellular carcinoma cells by activating the JAK2/STAT3/Snail pathway. Int J Oncol. 46:587–596. 2015.

15 

Yu J, Ren X, Chen Y, Liu P, Wei X, Li H, Ying G, Chen K, Winkler H and Hao X: Dysfunctional activation of neurotensin/IL-8 pathway in hepatocellular carcinoma is associated with increased inflammatory response in microenvironment, more epithelial mesenchymal transition in cancer and worse prognosis in patients. PLoS One. 8:e560692013. View Article : Google Scholar : PubMed/NCBI

16 

Bauerle KT, Schweppe RE, Lund G, Kotnis G, Deep G, Agarwal R, Pozdeyev N, Wood WM and Haugen BR: Nuclear factor κB-dependent regulation of angiogenesis, and metastasis in an in vivo model of thyroid cancer is associated with secreted interleukin-8. J Clin Endocrinol Metab. 99:E1436–E1444. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Birchmeier W and Behrens J: Cadherin expression in carcinomas: Role in the formation of cell junctions and the prevention of invasiveness. Biochim Biophys Acta. 1198:11–26. 1994.PubMed/NCBI

18 

Georgolios A, Batistatou A, Manolopoulos L and Charalabopoulos K: Role and expression patterns of E-cadherin in head and neck squamous cell carcinoma (HNSCC). J Exp Clin Cancer Res. 25:5–14. 2006.PubMed/NCBI

19 

Zhao Z, Ge J, Sun Y, Tian L, Lu J, Liu M and Zhao Y: Is E-cadherin immunoexpression a prognostic factor for head and neck squamous cell carcinoma (HNSCC)? A systematic review and meta-analysis. Oral Oncol. 48:761–767. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Jin B and Robertson KD: DNA methyltransferases, DNA damage repair, and cancer. Adv Exp Med Biol. 754:3–29. 2013. View Article : Google Scholar :

21 

Marsit CJ, Posner MR, McClean MD and Kelsey KT: Hypermethylation of E-cadherin is an independent predictor of improved survival in head and neck squamous cell carcinoma. Cancer. 113:1566–1571. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Shargh SA, Sakizli M, Khalaj V, Movafagh A, Yazdi H, Hagigatjou E, Sayad A, Mansouri N, Mortazavi-Tabatabaei SA and Khorram Khorshid HR: Downregulation of E-cadherin expression in breast cancer by promoter hypermethylation and its relation with progression and prognosis of tumor. Med Oncol. 31:2502014. View Article : Google Scholar : PubMed/NCBI

23 

Wang G, Hu X, Lu C, Su C, Luo S and Luo ZW: Promoter-hypermethylation associated defective expression of E-cadherin in primary non-small cell lung cancer. Lung Cancer. 62:162–172. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Li G, Liu Y, Yin H, Zhang X, Mo X, Tang J and Chen W: E-cadherin gene promoter hypermethylation may contribute to the risk of bladder cancer among Asian populations. Gene. 534:48–53. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Jin B, Ernst J, Tiedemann RL, Xu H, Sureshchandra S, Kellis M, Dalton S, Liu C, Choi JH and Robertson KD: Linking DNA methyltransferases to epigenetic marks and nucleosome structure genome-wide in human tumor cells. Cell Rep. 2:1411–1424. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Estève PO, Chang Y, Samaranayake M, Upadhyay AK, Horton JR, Feehery GR, Cheng X and Pradhan S: A methylation and phosphorylation switch between an adjacent lysine and serine determines human DNMT1 stability. Nat Struct Mol Biol. 18:42–48. 2011. View Article : Google Scholar :

27 

Hodge DR, Cho E, Copeland TD, Guszczynski T, Yang E, Seth AK and Farrar WL: IL-6 enhances the nuclear translocation of DNA cytosine-5-methyltransferase 1 (DNMT1) via phosphorylation of the nuclear localization sequence by the AKT kinase. Cancer Genomics Proteomics. 4:387–398. 2007.

28 

Shao N, Lu Z, Zhang Y, Wang M, Li W, Hu Z, Wang S and Lin Y: Interleukin-8 upregulates integrin β3 expression and promotes estrogen receptor-negative breast cancer cell invasion by activating the PI3K/Akt/NF-κB pathway. Cancer Lett. 364:165–172. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Bi LK, Zhou N, Liu C, Lu FD, Lin TX, Xuan XJ, Jiang C, Han JL, Huang H, Zhang CX, et al: Kidney cancer cells secrete IL-8 to activate Akt and promote migration of mesenchymal stem cells. Urol Oncol. 32:607–612. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Wang L, Tang C, Cao H, Li K, Pang X, Zhong L, Dang W, Tang H, Huang Y, Wei L, et al: Activation of IL-8 via PI3K/Akt-dependent pathway is involved in leptin-mediated epithelial-mesenchymal transition in human breast cancer cells. Cancer Biol Ther. 16:1220–1230. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Qian CN, Berghuis B, Tsarfaty G, Bruch M, Kort EJ, Ditlev J, Tsarfaty I, Hudson E, Jackson DG, Petillo D, et al: Preparing the ‘soil’: The primary tumor induces vasculature reorganization in the sentinel lymph node before the arrival of metastatic cancer cells. Cancer Res. 66:10365–10376. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Li MQ, Luo XZ, Meng YH, Mei J, Zhu XY, Jin LP and Li DJ: CXCL8 enhances proliferation and growth and reduces apoptosis in endometrial stromal cells in an autocrine manner via a CXCR1-triggered PTEN/AKT signal pathway. Hum Reprod. 27:2107–2116. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Di Croce L and Pelicci PG: Tumour-associated hypermethylation: Silencing E-cadherin expression enhances invasion and metastasis. Eur J Cancer. 39:413–414. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Grivennikov SI, Greten FR and Karin M: Immunity, inflammation, and cancer. Cell. 140:883–899. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Palena C, Hamilton DH and Fernando RI: Influence of IL-8 on the epithelial-mesenchymal transition and the tumor microenvironment. Future Oncol. 8:713–722. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Feinberg A: DNA methylation in cancer: Three decades of discovery. Genome Med. 6:362014. View Article : Google Scholar : PubMed/NCBI

37 

Schraufstatter IU, Chung J and Burger M: IL-8 activates endothelial cell CXCR1 and CXCR2 through Rho and Rac signaling pathways. Am J Physiol Lung Cell Mol Physiol. 280:L1094–L1103. 2001.PubMed/NCBI

38 

Lane HC, Anand AR and Ganju RK: Cbl and Akt regulate CXCL8-induced and CXCR1- and CXCR2-mediated chemotaxis. Int Immunol. 18:1315–1325. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Ochiai A: Dysfunction of cadherin cell adhesion system in cancer invasion and metastasis. Gan To Kagaku Ryoho. 26:565–571. 1999.In Japanese. PubMed/NCBI

40 

Zheng Z, Pan J, Chu B, Wong YC, Cheung AL and Tsao SW: Downregulation and abnormal expression of E-cadherin and beta-catenin in nasopharyngeal carcinoma: Close association with advanced disease stage and lymph node metastasis. Hum Pathol. 30:458–466. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 48 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang R, Peng L, Yang J, Zheng L, Xie P, Wang M, Huang B, Zhao H, Bao Y, Qian C, Qian C, et al: IL-8 suppresses E-cadherin expression in nasopharyngeal carcinoma cells by enhancing E-cadherin promoter DNA methylation. Int J Oncol 48: 207-214, 2016
APA
Zhang, R., Peng, L., Yang, J., Zheng, L., Xie, P., Wang, M. ... Qian, C. (2016). IL-8 suppresses E-cadherin expression in nasopharyngeal carcinoma cells by enhancing E-cadherin promoter DNA methylation. International Journal of Oncology, 48, 207-214. https://doi.org/10.3892/ijo.2015.3226
MLA
Zhang, R., Peng, L., Yang, J., Zheng, L., Xie, P., Wang, M., Huang, B., Zhao, H., Bao, Y., Qian, C."IL-8 suppresses E-cadherin expression in nasopharyngeal carcinoma cells by enhancing E-cadherin promoter DNA methylation". International Journal of Oncology 48.1 (2016): 207-214.
Chicago
Zhang, R., Peng, L., Yang, J., Zheng, L., Xie, P., Wang, M., Huang, B., Zhao, H., Bao, Y., Qian, C."IL-8 suppresses E-cadherin expression in nasopharyngeal carcinoma cells by enhancing E-cadherin promoter DNA methylation". International Journal of Oncology 48, no. 1 (2016): 207-214. https://doi.org/10.3892/ijo.2015.3226