Open Access

Na+/H+ exchanger regulatory factor 1 overexpression suppresses the malignant phenotype of MIAPaCa-2 pancreatic adenocarcinoma cells by downregulating Akt phosphorylation

  • Authors:
    • Xibo Zhang
    • Junjian Liu
    • Zhonglian Li
  • View Affiliations

  • Published online on: March 16, 2018     https://doi.org/10.3892/ol.2018.8289
  • Pages: 7725-7729
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Na+/H+ exchanger regulatory factor 1 (NHERF1) is reported to be associated with the development of numerous types of tumor; however, its effects on the metastasis of pancreatic adenocarcinoma are not fully understood. In the present study, it was revealed that the expression level of NHERF1 in pancreatic adenocarcinoma is decreased compared with normal pancreatic tissue based on the analysis of a protein expression database. The present study was undertaken in order to investigate the potential effects of NHERF1 overexpression on the malignant phenotype of MIAPaCa‑2 pancreatic adenocarcinoma cells. NHERF1 was stably overexpressed in this cell line, and Cell Counting Kit‑8, wound healing and Transwell assays were used to detect the proliferative and migratory abilities of the cells. NHERF1 overexpression suppressed proliferation in the MIAPaCa‑2 cell line compared with empty vector‑transfected (negative control) cells. Additionally, NHERF1 overexpression significantly inhibited the migration of MIAPaCa‑2 cells. The results of a western blot analysis identified that NHERF1 overexpression markedly decreased the expression of phosphorylated‑protein kinase B (p‑Akt), while no significant difference was observed between untransfected and negative control cells. Taken together, these results suggested that NHERF1 may be able to inhibit the proliferation and migration and alter the malignant phenotype of pancreatic adenocarcinoma cells via reduction of p‑Akt levels. These findings indicate a potential novel approach to the treatment of pancreatic adenocarcinoma.

Introduction

Pancreatic adenocarcinoma is the fourth leading cause of cancer-associated mortality in the USA, and pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease with a high mortality rate (1). The majority of patients are diagnosed at a late stage and, despite recent advances in chemotherapeutic approaches, the prognosis of pancreatic adenocarcinoma is extremely poor compared with other types of cancer (2,3). Tumor growth and migration are largely responsible for the high mortality rate of patients with pancreatic adenocarcinoma; therefore, it is important for researchers to investigate methods for preventing pancreatic adenocarcinoma cell proliferation and migration in order to improve the treatment of this disease (3,4).

Na+/H+ exchanger regulatory factor 1 (NHERF1; also known as sodium-hydrogen antiporter 3 regulator 1 or ERM-binding protein 50) is a multi-functional scaffolding protein that has different functions in a variety of types of cancer through its interactions with oncogenic or tumor-suppressive proteins (4,5). In breast cancer, NHERF1 has been demonstrated to inhibit proliferation by influencing the transduction of growth signals induced by epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor, and by modulating the expression of phosphatase and tensin homolog (6,7). By contrast, in prostate cancer, the expression of NHERF1 has been demonstrated to be increased, suggesting that NHERF1 may be associated with the carcinogenic potential of this cancer type (8). However, the function of NHERF1 in the proliferation and migration of pancreatic adenocarcinoma cells remains unresolved.

The objective of the present study was to determine the effects of NHERF1 expression on proliferative and migratory abilities of pancreatic adenocarcinoma cells by overexpressing NHERF1 in MIAPaCa-2 cells. The results revealed that NHERF1 may be able to inhibit the proliferative and migratory abilities of pancreatic adenocarcinoma cells by downregulating the phosphorylation of protein kinase B (Akt).

Materials and methods

Immunohistochemical (IHC) data, plasmids and cell lines

The IHC-based protein expression data, including high-resolution images, were downloaded from the Human Protein Atlas web portal, using NHERF1 as the search term (pancreatic cancer database; www.proteinatlas.org). All IHC images from pancreatic cancer and normal tissues were collected, and the sum of the integrated optical density (IOD) values of images were analyzed using ImagePro Plus software (version 6.0; Media Cybernetics, Inc., Rockville, MD, USA). The mean IOD values of these images were counted, which reflected the relative NHERF1 expression level in pancreatic cancer and normal pancrease tissues, respectively. The pBK-CMV-HA-NHERF1 wild-type (wt) plasmid and the empty vector plasmid (pBK-CMV-HA) were designed and synthesized by Sangon Biotech Co., Ltd. (Shanghai, China), and G418 resistance was encoded by the plasmid. MIAPaCa-2 human PDAC cells were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA).

Identification of stably transfected cells

MIAPaCa-2 cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C in an incubator with 5% CO2. DMEM was supplemented with 10% fetal bovine serum (Thermo Fisher Scientific, Inc.) and 1% penicillin-streptomycin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). For stable overexpression of NHERF1, MIAPaCa-2 cells were transfected with 2 µg pBK-CMV-HA-NHERF1 wt plasmid or the pBK-CMV-HA plasmid (negative control) using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Stably transfected cells were selected using 300 µg/ml G418 (Amresco, LLC, Solon, OH, USA) for 4 weeks and then maintained in maintenance culture medium containing G418 (150 µg/ml).

Western blotting

Cells were collected and total protein was extracted from cells stably expressing NHERF1 and from empty vector-transfected cells using radioimmunoprecipitation lysis buffer (Beijing CoWin Biotech Co., Ltd., Beijing, China) containing Halt™ Protease and Phosphatase Inhibitor Cocktail (100X; Thermo Fisher Scientific, Inc.). Protein levels were quantified using bicinchoninic acid assays (Beijing CoWin Biotech Co., Ltd.). Subsequently, 30 µg protein from each sample was subjected to SDS-PAGE (10% gel). Proteins were then transferred to nitrocellulose membranes (Sigma-Aldrich; Merck KGaA). The membranes were blocked with 5% skimmed milk (dissolved in TBST) for 1 h at 25°C, prior to incubation with rabbit anti-human primary antibodies against NHERF1 (1:1,000 dilution, cat. no. ab88238), and GAPDH (1:5,000 dilution, cat. no. ab70699) (both from Abcam, Cambridge, UK), Akt (1:1,000 dilution, cat. no. 9272; Cell Signaling Technology, Inc., Danvers, MA, USA) or phospho-Akt (p-AKT) (phospho-Ser473, 1:2,000 dilution, cat. no. 4060; Cell Signaling Technology, Inc.) overnight at 4°C, followed by incubation with goat anti-rabbit horseradish peroxidase-conjugated secondary antibodies (1:1,000 dilution, cat. no. ab6721; Abcam) for 1 h at room temperature. Detection was facilitated using an enhanced chemiluminescence western blot kit (Beijing CoWin Biotech Co., Ltd.) and images were analyzed using ImageJ software (version 1.62; National Institutes of Health, Bethesda, MD, USA).

Cell proliferation assay

Cells stably expressing NHERF1 were plated at a density of 5×103 cells/well in 96-well plates at 37°C in an incubator with 5% CO2. Cell proliferation was then assessed every 24 h for 96 h using a Cell Counting Kit-8 (CCK-8; Sigma-Aldrich; Merck KGaA) according to the manufacturer's instructions. For each sample at each time-point, 6 wells were analyzed, and the experiment was repeated independently three times.

Wound healing assay

Cells were plated at 3×105 cells/well in 6-well plates and grown to 100% confluence. Scratches were created with 1-ml pipette tips in the cell monolayer, and an image was immediately captured (0 h). Subsequent images were captured every 12 h, and the migration (scratch width) relative to 0 h was calculated using Image-Pro Plus analysis software (version 6.0; Media Cybernetics).

Transwell assay

For the Transwell assay, DMEM containing 10% fetal bovine serum was added to the lower chamber of Transwell culture plates. Subsequently, three groups of cells (untransfected, empty vector and NHERF1-overexpressing) were seeded into the upper chambers of Transwell 24-well culture plates in serum-free DMEM. Following incubation for 24 h at 37°C, the cells on the upper membrane were removed with a cotton swab, and the cells that had migrated through the membrane were fixed in 4% paraformaldehyde for 15 min at 25°C, and stained with 0.5% crystal violet for 15 min at 25°C. The mean number of cells that had traversed the membrane was calculated in five random fields under a light microscope (magnification, ×400).

Statistical analysis

All experiments were repeated at least three times. SPSS software (version 21.0; IBM Corp., Armonk, NY, USA) and GraphPad Prism 5 (GraphPad Software, Inc., La Jolla, CA, USA) were used to analyze data. IHC data were analyzed using an independent samples t-test. Growth curves, Transwell assay and wound healing assay results were analyzed using a repeated-measures analysis of variance with Fisher's least significant difference post hoc tests. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of NHERF1 is downregulated in pancreatic adenocarcinoma tissues

To identify the expression level of NHERF1 in pancreatic adenocarcinoma tissues, sum of the IOD values of NHERF1 expression levels in pancreatic adenocarcinoma tissues (n=3) and adjacent normal pancreatic tissues (n=11) were compared using a Human Protein Atlas Database dataset. The results confirmed that the expression of NHERF1 was significantly downregulated in pancreatic adenocarcinoma tissues compared with adjacent tissues (P<0.01; Fig. 1), indicating that decreased NHERF1 expression may be associated with pancreatic cancer progression.

Generation of PDAC cells overexpressing NHERF1

As presented in Fig. 2, NHERF1 was overexpressed in MIAPaCa-2 cells transfected with the NHERF1 plasmid compared with the cells transfected with the empty vector. These cells were used to verify whether NHERF1 is able to attenuate the malignant phenotype in subsequent experiments. Cells transfected with NHERF1 were designated MIA-NHERF1 and cells transfected with the empty vector were designated MIA-vector.

NHERF1 overexpression inhibits the proliferative ability of MIAPaCa-2 cells

The ability of NHERF1 to modulate the proliferation of PDAC cells was analyzed using a CCK-8 assay. The results suggested that NHERF1 overexpression significantly inhibited the proliferative capacity of MIAPaCa-2 cells following 48 h of incubation, compared with that of control cells (P<0.05; Fig. 3). There were no significant differences observed between MIA-vector and untransfected MIAPaCa-2 cells.

NHERF1 overexpression inhibits the migratory ability of MIAPaCa-2 cells

A proliferation assay indicated that NHERF1 exhibited the ability to inhibit tumor cell proliferation. Since proliferation and migration are closely associated with tumor progression (9), the ability of NHERF1 to suppress cell migratory ability was investigated. The wound-healing assay revealed that, after 24 h, the wound width was significantly decreased in the MIA-vector and untransfected MIAPaCa-2 group, while the width reduction was significantly inhibited in the MIA-NHERF1 group (P<0.05), as presented in Fig. 4A. Consistently, in the Transwell assay, NHERF1 overexpression significantly decreased the number of migrated cells compared with the vector control group (P<0.05) (Fig. 4B). These results suggested that NHERF1 overexpression was able to inhibit the migratory abilities of MIAPaCa-2 cells.

NHERF1 overexpression reduces the Akt phosphorylation in MIAPaCa-2 cells

The activation of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway contributes to cell growth and survival of numerous types of cancer (10,11), including pancreatic adenocarcinoma (12), and NHERF1 has been demonstrated to regulate the PI3K/Akt signaling pathway in breast cancer (13). Thus, the activation of Akt was examined in the present study. The results identified markedly decreased levels of p-Akt in the MIA-NHERF1 group compared with the Mia-vector group, whereas there was no notable difference between the MIA-vector and untransfected MIAPaCa-2 cells. These results suggest that NHERF1 may inhibit Akt phosphorylation in pancreatic cancer cells (Fig. 5).

Discussion

In the present study, it was demonstrated that the expression level of NHERF1 was downregulated in pancreatic adenocarcinoma tissues, and that NHERF1 overexpression may inhibit the proliferative and migratory abilities of MIAPaCa-2 PDAC cells in vitro, while downregulating Akt phosphorylation. Therefore, NHERF1 may represent a metastasis-suppressing protein in pancreatic adenocarcinoma. The data suggest that NHERF1 expression may be able to inhibit the malignant phenotype of pancreatic adenocarcinoma cells via downregulating the expression of p-Akt.

Abnormalities in NHERF1 expression have been demonstrated to be associated with the occurrence, development and metastasis of cancer (6,14). NHERF1 is hypothesized to directly or indirectly affect adenocarcinoma behaviors via interaction with other proteins and signal transduction (15). NHERF1 is known to form a protein complex with EGFR (16), thereby mediating the internalization and signal transduction of EGFR to regulate oncogenic processes. NHERF1 also serves as a binding partner for G protein-coupled estrogen receptor (GPER), and its overexpression promotes the stability and activation of GPER in estrogen receptor-positive invasive breast cancer (17). Furthermore, a previous report outlined a complex function of NHERF1 in intestinal morphology and presented evidence for its in vivo tumor-suppressive function upstream of the Wnt-β-catenin and Hippo-YAP signaling pathways (18). However, the physiological function of NHERF1 in pancreatic cancer has largely remained unresolved.

Components of the PI3K/Akt/mTOR signaling pathway are commonly upregulated in malignant tumors, and increased Akt phosphorylation is commonly observed (19). The activation of Akt signaling is associated with cell proliferation, migration and invasion (20). Notably, it has been reported that NHERF1 inhibits the migration and invasion of human breast cancer cells via the PI3K/Akt signaling pathway (13). However, it remains unknown known whether or not NHERF1 may be able to attenuate the malignant phenotype of pancreatic cancer cells via inhibition of Akt phosphorylation.

In pancreatic adenocarcinoma tissues, NHERF1 exhibited relatively low endogenous expression. NHERF1 expression was increased using stable transfection with a pBK-CMV-HA-NHERF1 wt plasmid. Analysis indicated that NHERF1 overexpression inhibited the proliferation of MIAPaCa-2 cells, and also suppressed cell migration. Cell proliferation and migration are associated with tumor development and substantially contribute to the mortality of patients with tumors (24). Thus, the results of the present study are relevant to the understanding of tumor development and therapeutics. With regard to the molecular mechanism underlying the inhibition of cell proliferation and migration by NHERF1, two possible mechanisms are hypothesized. The first is that NHERF1 interacts with molecular partners, which serve important functions in proliferation and migration; for example, NHERF1 is reported to interact with EGFR and regulate EGFR signaling (21). The other hypothesis is that NHERF1 may be associated with the downregulation of Akt phosphorylation. Further experimental studies are required to confirm these hypotheses, and this may be a focus of future study.

NHERF1 participates in cell signaling and has multiple physiological functions. The results of the present study demonstrate that NHERF1 can regulate the malignant behaviors (proliferative and migratory abilities) of MIAPaCa-2 PDAC cells by downregulating Akt phosphorylation, and support that NHERF1 may function as a metastasis-suppressing protein in pancreatic adenocarcinoma. Thus, NHERF1 may be a potential therapeutic target for the treatment of pancreatic adenocarcinoma.

Glossary

Abbreviations

Abbreviations:

NHERF1

Na+/H+ exchanger regulatory factor 1

PDAC

pancreatic ductal adenocarcinoma

EGFR

epidermal growth factor receptor

PI3K

phosphatidylinositol 3-kinase

Akt

protein kinase B

References

1 

Mann KM, Ying H, Juan J, Jenkins NA and Copeland NG: KRAS-related proteins in pancreatic cancer. Pharmacol Ther. 168:29–42. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Vijayvergia N and Cohen SJ: Personalized medicine in sporadic pancreatic cancer without homologous recombination-deficiency: Are we any closer? J Gastrointest Oncol. 7:727–737. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Miyoshi E and Kamada Y: Application of glycoscience to the early detection of pancreatic cancer. Cancer Sci. 107:1357–1362. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Cheng S, Li Y, Yang Y, Feng D, Yang L, Ma Q, Zheng S, Meng R, Wang S, Wang S, et al: Breast cancer-derived K172N, D301V mutations abolish Na+/H+ exchanger regulatory factor 1 inhibition of platelet-derived growth factor receptor signaling. FEBS Lett. 587:3289–3295. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Shibata T, Chuma M, Kokubu A, Sakamoto M and Hirohashi S: EBP50, a beta-catenin-associating protein, enhances Wnt signaling and is over-expressed in hepatocellular carcinoma. Hepatology. 38:178–186. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Fraenzer JT, Pan H, Minimo L Jr, Smith GM, Knauer D and Hung G: Overexpression of the NF2 gene inhibits schwannoma cell proliferation through promoting PDGFR degradation. Int J Oncol. 23:1493–1500. 2003.PubMed/NCBI

7 

Yao W, Feng D, Bian W, Yang L, Li Y, Yang Z, Xiong Y, Zheng J, Zhai R and He J: EBP50 inhibits EGF-induced breast cancer cell proliferation by blocking EGFR phosphorylation. Amino Acids. 43:2027–2035. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Ma Q, Jiao Y, Hao Y, Yan S, Lyu N, Gao H, Li D, Liu Q, Zheng J and Song N: Targeting of NHERF1 through RNA interference inhibits the proliferation and migration of metastatic prostate cancer cells. Oncol Lett. 11:1149–1154. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Chen X, Yang H, Zhang S, Wang Z, Ye F, Liang C, Wang H and Fang Z: A novel splice variant of supervillin, SV5, promotes carcinoma cell proliferation and cell migration. Biochem Biophys Res Commun. 482:43–49. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Talbert EE, Yang J, Mace TA, Farren MR, Farris AB, Young GS, Elnaggar O, Che Z, Timmers CD, Rajasekera P, et al: Dual inhibition of MEK and PI3K/Akt rescues cancer cachexia through both tumor extrinsic and intrinsic activities. Mol Cancer Ther. 16:344–356. 2016. View Article : Google Scholar : PubMed/NCBI

11 

McCubrey JA, Rakus D, Gizak A, Steelman LS, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Yang LV, Montalto G, Cervello M, et al: Effects of mutations in Wnt/beta-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-diverse effects on cell growth, metabolism and cancer. Biochim Biophys Acta. 1863:2942–2976. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Rumman M, Jung KH, Fang Z, Yan HH, Son MK, Kim SJ, Kim J, Park JH, Lim JH and Hong S: HS-173, a novel PI3K inhibitor suppresses EMT and metastasis in pancreatic cancer. Oncotarget. 7:78029–78047. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Li H, Zhang B, Liu Y and Yin C: EBP50 inhibits the migration and invasion of human breast cancer cells via LIMK/cofilin and the PI3K/Akt/mTOR/MMP signaling pathway. Med Oncol. 31:1622014. View Article : Google Scholar : PubMed/NCBI

14 

Bellizzi A, Greco MR, Rubino R, Paradiso A, Forciniti S, Zeeberg K, Cardone RA and Reshkin SJ: The scaffolding protein NHERF1 sensitizes EGFR-dependent tumor growth, motility and invadopodia function to gefitinib treatment in breast cancer cells. Int J Oncol. 46:1214–1224. 2015. View Article : Google Scholar : PubMed/NCBI

15 

James MF, Beauchamp RL, Manchanda N, Kazlauskas A and Ramesh V: A NHERF binding site links the betaPDGFR to the cytoskeleton and regulates cell spreading and migration. J Cell Sci. 117:2951–2961. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Curto M, Cole BK, Lallemand D, Liu CH and McClatchey AI: Contact-dependent inhibition of EGFR signaling by Nf2/Merlin. J Cell Biol. 177:893–903. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Meng R, Qin Q, Xiong Y, Wang Y, Zheng J, Zhao Y, Tao T, Wang Q, Liu H, Wang S, et al: NHERF1, a novel GPER associated protein, increases stability and activation of GPER in ER-positive breast cancer. Oncotarget. 7:54983–54997. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Georgescu MM, Gagea M and Cote G: NHERF1/EBP50 suppresses Wnt-beta-catenin pathway-driven intestinal neoplasia. Neoplasia. 18:512–523. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Lim HJ, Wang X, Crowe P, Goldstein D and Yang JL: Targeting the PI3K/PTEN/AKT/mTOR pathway in treatment of sarcoma cell lines. Anticancer Res. 36:5765–5771. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Xiao J, Lin HY, Zhu YY, Zhu YP and Chen LW: miR-126 regulates proliferation and invasion in the bladder cancer BLS cell line by targeting the PIK3R2-mediated PI3K/Akt signaling pathway. Onco Targets Ther. 9:5181–5193. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Peng Z, Wang Q, Zhang Y, He J and Zheng J: EBP50 interacts with EGFR and regulates EGFR signaling to affect the prognosis of cervical cancer patients. Int J Oncol. 49:1737–1745. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2018
Volume 15 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang X, Liu J and Li Z: Na+/H+ exchanger regulatory factor 1 overexpression suppresses the malignant phenotype of MIAPaCa-2 pancreatic adenocarcinoma cells by downregulating Akt phosphorylation. Oncol Lett 15: 7725-7729, 2018
APA
Zhang, X., Liu, J., & Li, Z. (2018). Na+/H+ exchanger regulatory factor 1 overexpression suppresses the malignant phenotype of MIAPaCa-2 pancreatic adenocarcinoma cells by downregulating Akt phosphorylation. Oncology Letters, 15, 7725-7729. https://doi.org/10.3892/ol.2018.8289
MLA
Zhang, X., Liu, J., Li, Z."Na+/H+ exchanger regulatory factor 1 overexpression suppresses the malignant phenotype of MIAPaCa-2 pancreatic adenocarcinoma cells by downregulating Akt phosphorylation". Oncology Letters 15.5 (2018): 7725-7729.
Chicago
Zhang, X., Liu, J., Li, Z."Na+/H+ exchanger regulatory factor 1 overexpression suppresses the malignant phenotype of MIAPaCa-2 pancreatic adenocarcinoma cells by downregulating Akt phosphorylation". Oncology Letters 15, no. 5 (2018): 7725-7729. https://doi.org/10.3892/ol.2018.8289