Proton pump inhibitor pantoprazole inhibits the proliferation, self‑renewal and chemoresistance of gastric cancer stem cells via the EMT/β‑catenin pathways

  • Authors:
    • Shuitu Feng
    • Zhigao Zheng
    • Lihua Feng
    • Lihong Yang
    • Zuhong Chen
    • Yubiao Lin
    • Yingqin Gao
    • Yide Chen
  • View Affiliations

  • Published online on: October 7, 2016     https://doi.org/10.3892/or.2016.5154
  • Pages: 3207-3214
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The cancer stem cell (CSC) model suggests that a small subset of cancer cells possess stem cell properties and plays a crucial role in tumor initiation, metastasis and resistance to anticancer therapy. Exploration of the specific therapies targeting at CSCs has been a crucial issue in antitumor research. Gastric cancer (GC) cells often exist in an ischemic microenvironment with acidic conditions in vivo, thus maintenance of cellular pH homeostasis is important for the survival and function of GC cells. Proton pump inhibitors (PPIs) may prevent intracellular proton extrusions which consequently reduce cancer cell survival under acidic conditions. The effects of PPIs on the suppression of the viability and invasiveness of GC cells have been reported, but the functional role of pantoprazole (PPZ) in GC cells remains unknown. In this study, we found that when cells were treated with PPZ, the 5‑fluorouracil (5‑FU) chemosensitivity was upregulated, meanwhile the sphere formation ability and the relative expression levels of stem cell markers CD44, CD24, ABCG2, EpCAM and Lgr5 were significantly decreased. It was hypothesized that PPZ inhibits the GC CSCs. Successively a sphere formation culture was performed to establish CSC models and the effect of PPZ on GC CSCs from SGC-7901 and HGC‑27 cells was explored. The addition of PPZ reduced the relative expression of CSC markers and anti‑drug markers accompanied by a decrease in proliferation, 5‑FU chemoresistance and self‑renewal capacity via epithelial‑mesenchymal transition (EMT)/β‑catenin pathways. The study suggests that PPZ could be a promising novel specific therapeutic strategy for targeting GC CSCs.

Introduction

Gastric cancer (GC) is one of the most common malignancies and a leading cause of cancer-related deaths worldwide (1) which remains prevalent in Eastern Asia, South America and Eastern Europe (2,3). Common treatments for advanced GC include surgery, chemotherapy and radiotherapy. Although chemotherapy is effective for suppressing cancer progression and prolonging survival to some extent, chemoresistance represents a predominant obstacle towards the chemotherapeutic treatment of GC (4). It is worth noting that an extremely efficient mechanism of tumor resistance to drugs is the proton pump-mediated acidification of the tumor microenvironment (5).

GC cells often exist in an ischemic microenvironment with acidic conditions which is a consequence of the production of acidic by-products from explosive glycolysis in vivo (6,7). Maintenance of cellular pH homeostasis is vital for the survival and function of cancer cells (8,9). Hydrogen/potassium adenosine triphosphatase (H+/K+-ATPase) plays a vital role in the maintenance of cellular pH homeostasis by exchanging luminal K+ for cytoplasmic H+. This proton pump also participates in the formation of abnormal pH gradients that are typical of GC cells during tumorigenesis (10). Proton pump inhibitors (PPIs) inhibit gastric H+/K+-ATPase irreversibly which prevents intracellular proton extrusions in GC cells consequently reducing cancer cell survival under acidic conditions (11). It was reported that rabeprazole attenuates the cell viability of human GC cells through inactivation of the ERK1/2 signaling pathway (12). Moreover, pantoprazole (PPZ) could be used to suppress the invasiveness of SGC-7901/ADR cells by targeting epithelial-mesenchymal transition (EMT) (13).

Cancer stem cells (CSCs) play pivotal roles in cancer initiation, progression, recurrence and chemoresistance (1417). CSCs give rise to tumors through self-renewal and are able to differentiate into multiple cell types (1821). Chemotherapies that kill the bulk of cancer cells, may ultimately fail as they do not eliminate CSCs that then cause the relapse of tumors (22). Recently, it has been established that CSCs are linked to EMT, metastasis, drug resistance, progression and relapse of GC (2326). As a result, exploitation of specific therapies targeting CSCs has been a crucial issue in the chemotherapeutic treatment of GC.

In the present study, we found that the PPI PPZ suppressed the proliferation, sphere formation and GC stem cell-mediated 5-fluorouracil (5-FU) chemoresistance of SGC-7901 and HGC-27 cells by targeting EMT and β-catenin signaling for the first time and our findings suggest that PPIs may serve as a promising CSC-oriented novel antineoplastic agent.

Materials and methods

Cell culture, sphere formation culture and reagents

Human GC cell lines SGC-7901 and HGC-27 were purchased from Auragene Bioscience Co. (Changsha, China). Under standard conditions, both cell lines were cultured in normal Dulbeccos modified Eagles medium (DMEM) (Gibco-BRL, Grand Island, NY, USA) with 10% fetal bovine serum and 100 U/ml penicillin at 37°C in a humidified atmosphere of 95% air and 5% CO2. Under sphere culture conditions, parental cell lines or the floating spheres obtained from transfections were plated in serum-free defined media (SFDM): low-glucose (1 g/l) DMEM supplemented with L-glutamine, sodium pyruvate, penicillin/streptomycin (Wisent, Inc.), 20 ng/ml basic FGF, 20 ng/ml EGF and B27 (Invitrogen, Grand Island, NY, USA) using 24-well ultra-low attachment plates (Corning, Inc., Tewksbury, MA, USA). Spheres were dissociated with trypsin every 5–7 days and split to 1:3 ratio for next sphere passage if it was necessary. PPI PPZ (H20010032) was obtained from Hangzhou Meidong Pharmaceutical Co., Ltd. (Hangzhou, China). SGC-7901 and HGC-27 parental cells or spheres were treated with 100 µg/ml PPZ to generate cell models of interest.

Flow cytometric analysis

Cells (1×106) were labeled with PE-conjugated anti-CD44 (BioLegend, Inc., San Diego, CA, USA) and FITC-conjugated CD24 (BD Pharmingen, Mississauga, ON, Canada) for 20 min, washed twice, resuspended in PBS and analyzed on a BD FACSCalibur™ platform (BD Biosciences). Data were analyzed by FCS Express software (BD Biosciences) using floating quadrants to enumerate negative, single- and double-positive populations.

Drug resistance assay

Parental cells and spheres of P4 were planted at 2,000 cells/well in 96-well plates and then divided into three groups as follows: treatment with 5-FU (20 µg/ml), treatment with pantoprazol (PPZ; 100 µg/ml) and co-treatment with 5-FU and PPZ (5-FU + PPZ). Cell viability was examined. Drug resistance was determined after treatment for 96 h by MTT assay.

Self-renewal assay

Sphere formation assay was performed to detect the self-renewal capacity. A total of 4,000 cells/well were seeded in ultra-low attachment 6-well plate (Corning, Inc.) in SFDM medium for 2 weeks, after which sphere formation was assessed by counting the number of spheres (≥3 cells) under a microscope.

Cell growth assay of parental cells and spheres

A total of 2,000 cells were plated in 96-well plates and cultured in a CO2 incubator. The cells were harvested at 24, 48, 72 and 96 h. The optical density at 570 nm (OD570) of each well was measured with an ELISA reader (ELx800; BioTek Instruments, Inc.).

Real-time RT-PCR

Total RNA was extracted from the cells with TRIzol reagent (Invitrogen) following the manufacturer's instructions. The relative mRNA levels of CSC markers of CD44, CD24, ABCG2, EpCAM, Lgr5 and drug-resistance markers BMI1, ALDH1, Tcf4 were detected by real-time RT-PCR using the standard SYBR-Green RT-PCR kit (Takara Bio, Inc., Otsu, Japan) following the manufacturer's instructions and β-actin was used as an internal control. The specific primer pairs are listed in Table I. The relative expression of target genes was quantified using GraphPad Prism 4.0 software (GraphPad Software, San Diego, CA, USA) and the 2−ΔΔCt method (27).

Table I.

The primer sequences used in real-time RT-PCR.

Table I.

The primer sequences used in real-time RT-PCR.

Gene Primer sequences
CD44Sense CATCCCAGACGAAGACAGTCC
Antisense TGATCAGCCATTCTGGAATTTG
CD24Sense GACATGGGCAGAGCAATGGTGGC
Antisense GAGTGAGACCACGAAGAGACTGGC
ABCG2Sense CTGAGATCCTGAGCCTTTGG
Antisense TGCCCATCACAACATCATCT
EpCAMSense CGCCATATGCAGGAAGAATGTGT
Antisense CGCCTCGAGTTATTTTAGACCCTGCATTG
Lgr5Sense CCCGGGTTTCAGAGACAACTTC
Antisense TCCACATGCTTTATTCCAGCAATC
BMI1Sense ACGATGCCCAGCAGCAATGACT
Antisense AAGTGGACCATTCCTTCTCCAGGT
ALDH1Sense GATGAAGCTGCGGAATTTG
Antisense TCTTTGCTCGTTCAATGCTC
Tcf4Sense TGCGATGTTTTCACCTCCTG
Antisense TGCCAAAGAAGTTGGTCCATT
β-actinSense AGGGGCCGGACTCGTCATACT
Antisense GGCGGCACCACCATGTACCCT
Western blot analysis

Cells were solubilized in cold RIPA lysis buffer. Subsequently, the proteins were separated with 8% SDS-PAGE and then transferred to a PVDF membrane. The membranes were blocked in 5% non-fat dried milk in PBST for 3 h and then incubated overnight with specific primary antibodies with β-actin as a control. After incubation with the goat anti-rabbit or anti-mouse secondary antibody, immune complexes were detected using an ECL kit (Auragene Bioscience Co.). The primary antibodies against CD44 (1:1,000, ab54037), CD24 (1:1,000, ab113289), ABCG2 (1:1,000, ab63907), EpCAM (1:1,000, ab32392), Lgr5 (1:1,000, ab75732), BMI1 (1:1,000, ab126783), ALDH1 (1:1,000, ab6192), Tcf4 (1:1,000, ab60727), E-cadherin (1:1,000, ab15148), N-cadherin (1:1,000, ab18203), vimentin (1:1,000, ab133260) and Snail (1:1,000, ab82846) were all obtained from Abcam (USA), and total β-catenin (1:1,000, #9562) and active β-catenin (1:1,000, #4270) were purchased from Cell Signaling Technology, Inc. (CST; USA) The images were captured using GeneSnap software from SynGene (Cambridge, UK). The protein levels were normalized to β-actin.

Statistical analysis

The data are shown as the mean ± SD. The Student's t-test was used to analyze the differences between the experimental and control groups. Statistical analyses were performed using SPSS 11.0 software (SPSS, Inc., Chicago, IL, USA), and P<0.05 was considered to indicate a statistically significant difference.

Results

PPZ enhances 5-FU chemosensitivity and suppresses sphere formation and the expression of stem cell markers in GC cell lines

In order to ascertain whether PPZ exerts synergistic action with 5-FU to inhibit cell proliferation, SGC-7901 and HGC-27 cells were divided into three groups: 5-FU-treated, PPZ-treated and 5-FU combined with PPZ-treated. The results showed that the inhibition of proliferation by PPZ appeared after 48 h and until 96 h, inhibition was still slight. After addition of 5-FU (24 h), inhibition of proliferation appeared; at 48 until 96 h, it became significant. When cells were co-treated with 5-FU + PPZ, the inhibition of proliferation was most obvious. This suggested that PPZ enhanced 5-FU chemosensitivity of both the SGC-7901 and HGC-27 cells (Fig. 1A). Then the sphere formation of SGC-7901 and HGC-27 cells was assessed and the results showed that after PPZ treatment, the self-renewal efficiency of both cell lines declined obviously, not only the sphere numbers but also the size (Fig. 1C). CSCs play a role in chemoresistance, thus real-time RT-PCR and western blot analysis were performed to detect the relative expression of stem cell markers. In both cell lines, after PPZ treatment, the mRNA levels and protein expression of CD44, CD24, ABCG2, EpCAM and Lgr5 were reduced significantly (Fig. 1B). All of these results suggest that the antitumor effect of PPZ may be related to the mediation of GC cell stemness.

The establishment of GC CSC models

Successive sphere formation culture was performed to establish the CSC models using SGC-7901 and HGC-27 cell lines. The relative mRNA levels of classic CSC marker genes were used as a measurement of the CSC proportion. It was found that the relative mRNA levels of classic CSC markers CD44, CD24, ABCG2, EpCAM and Lgr5 were significantly increased from P1 to P3, but there was no significant difference between P3 to P4 (Fig. 2A). These results suggest that when spheres were cultured to P4, the CSC proportion reached a relative peak amplitude using these methods. Flow cytometric analysis was used to detect the CD133- and CD24-positive cell ratio of parental cells and P4 spheres and the results showed that compared with the parental groups the ratio of CD133+/CD24+ cells was enhanced obviously in the P4 sphere groups (Fig. 2B). It was concluded that during the successive sphere formation culture to P4, the GC CSCs were effectively enriched and the P4 spheres were able to be used as the GC CSC models in the following experiments.

PPZ inhibits the expression of CSC markers and drug-resistance markers

P4 spheres of both SGC-7901 and HGC-27 cells were treated with PPZ (100 µg/ml), and the expression of CSC markers CD44, CD24, ABCG2, EpCAM, Lgr5 and drug-resistance markers BMI1, ALDH1 and Trf4 was detected. Not only CSC marker genes but also anti-drug genes were all significantly inhibited at both the mRNA and protein levels when compared with the spheres without PPZ treatment (Fig. 3). The results suggest that PPZ exerts its tumor inhibition effect by reducing the stemness of GC cells.

PPZ enhances 5-FU chemosensitivity, and suppresses cell proliferation and sphere formation of GC-initiating cells

P4 spheres of both SGC-7901 and HGC-27 cells were used as GC initiation cell models which were treated with PPZ (100 µg/ml). Then the cell proliferation was measured by MTT assay. Compared with the P4 spheres without PPZ, the cell proliferation capacity was obviously decreased after 48 h and became even more significant after 72 h in both cell lines (Fig. 4A). P4 spheres of both cell lines were treated with 10–50 µg/ml 5-FU combined with or without PPZ under normal culture conditions. The cell viability assay showed that the cell viability was significantly inhibited when cells were co-treated with 5-FU + PPZ which indicated that PPZ enhanced the 5-FU sensitivity of the GC-initiating cells at least to some extent (Fig. 4B). Then the sphere formation of both P4 spheres was measured. After PPZ treatment, the self-renewal efficiency of both sphere types was obviously declined, not only sphere numbers but also the size (Fig. 4C). These results demonstrated that PPZ could exert its tumor inhibitory effect by reducing the stemness of GC cells at the cell functional level.

PPZ suppresses expression of EMT-related genes and the activation of β-catenin signaling

To explore the potential downstream molecular pathways underlying the targeting of stemness inhibition by PPZ, we assessed the expression of genes involved in EMT and classic CSC-related signaling pathways including E-cadherin, N-cadherin, vimentin, Snail and total/active β-catenin by western blot analysis in both types of P4 spheres with or without PPZ treatment. A significant reduction in the expression of N-cadherin, vimentin, Snail and total/active β-catenin proteins and upregulation of E-cadherin were observed in the spheres treated with PPZ in both types of GC spheres (Fig. 5). It was demonstrated that the inhibitory effect associated with the PPZ targeting of GC CSCs was mediated by EMT and activation of the β-catenin signaling at least partly.

Discussion

Current antineoplastic strategies are aimed at promoting the efficiency and specificity of therapies for GC. Intracellular proton extrusion in GC cells has been reported to promote cancer cell survival under acidic conditions via H+/K+-ATPase. PPZ is a frequently used second-generation PPI that irreversibly inactivates gastric H+/K+-ATPase. Early as 2004, relative research found that PPI selectively induced in vivo and in vitro apoptotic cell death in GC and could be used for a selective anticancer effect (28).

In regards to GC chemotherapy, the results are not satisfactory, and among the tested chemotherapeutic agents, only a limited number of compounds [5-FU, adriamycin (doxorubicin) and cisplatin] have demonstrated response rates ranging from 15 to 50% selectively. Recent studies have elucidated the presence of CSCs that have the exclusive ability to regenerate tumors. These CSCs share many characteristics with normal stem cells, including self-renewal, differentiation and drug-resistance (29). The presence of CSCs has already been associated with chemotherapeutic failure in a variety of solid tumors including GC. Thus, revealing the underlying molecular mechanisms responsible for maintenance and chemoresistance of CSCs has become a crucial issue in the clinical treatment of GC. PPZ was reported to suppress proliferation and restore the chemosensitivity of GC SGC-7901 cells by inhibiting the STAT3 signaling pathway (30). It was found that PPZ can effectively reverse the aggressiveness and EMT marker expression of SGC-7901/ADR (adriamycin-resistant) cells and EMT was a typical feature of CSCs. Our research showed that associated with the proliferation, and 5-FU resistance inhibition, administration of PPZ decreased the expression of GC CSC markers. Thus, we hypothesized that PPZ could play a role in targeting CSCs.

The CSC model suggests that a small subset of cancer cells possesses stem cell properties and plays a crucial role in tumor initiation, metastasis and resistance to anticancer therapy (31). The characteristic features of CSCs include: i) self-renewing (tested by sphere formation in SFDM); ⅱ) high tumorigenicity in xenograft-based model; and ⅲ) ability to differentiate to the cell types of the tumor of origin (32,33). To date, although the purity is not 100%, there are three relatively satisfactory methods by which to establish CSC models in tumor cells: side population isolation, sphere culture and immunomagnetic bead isolation. No matter what isolation method is used, the relative expression levels of stem cell surface markers are favourable indices to measure the abundance of CSC models. In our study, we adopted a successive sphere culture method to establish our GC CSC models. Using qPCR, we found that the relative mRNA levels of stem cell markers were significantly upregulated from P1 to P3, but were not further upregulated from P3 to P4. The results suggested that, using the successive sphere culture method, we received a comparatively high proportion of GC CSCs from P4. Thus, in our following experiments, the P4 spheres were used as our CSC models.

Upon pre-treatment with PPI, ERK1/2 phosphorylation was completely inhibited; the inhibitory action on the phosphorylation of ERK1/2 might contribute to the induction of apoptosis in GC cells by PPI (34). Furthermore, PPZ treatment resulted in a profound reduction in both total and phosphorylated forms of Akt and GSK-3β, which in turn suppressed the adriamycin-induced Wnt/β-catenin signaling in SGC-7901/ADR cells. It is possible to suppress the invasiveness of SGC-7901/ADR cells by PPZ which targets the EMT and Akt/GSK-3β/β-catenin signaling (13). It is worth mentioning that EMT and Wnt/β-catenin signaling are classic relative signaling pathways that mediate the stemness of CSCs. EMT is an important way to induce CSC formation in a number of solid tumors. Moreover, EMT is associated with increased expression of stem cell-related transcription factors and with increased tumorigenic ability (31). In our experiment, when spheres were treated with PPZ, the EMT course and the activation of β-catenin were both inhibited to some extent which demonstrated that PPZ exerts its antitumor effect by targeting CSCs via the EMT/β-catenin pathways.

Our research for the first time demonstrated that PPZ could be used to promote a selective anticancer effect targeting GC CSC inhibition, but the underlying molecular mechanism should be further explored. PPZ may be a promising breakthrough in GC therapy.

Acknowledgements

This study was supported by the Science and Technology Program of Haicang District of Xiamen City (grant no. 350205Z20144010-05).

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Center MM, DeSantis C and Ward EM: Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomarkers Prev. 19:1893–1907. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Smith MG, Hold GL, Tahara E and El-Omar EM: Cellular and molecular aspects of gastric cancer. World J Gastroenterol. 12:2979–2990. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Wöhrer SS, Raderer M and Hejna M: Palliative chemotherapy for advanced gastric cancer. Ann Oncol. 15:1585–1595. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Spugnini EP, Buglioni S, Carocci F, Francesco M, Vincenzi B, Fanciulli M and Fais S: High dose lansoprazole combined with metronomic chemotherapy: A phase I/II study in companion animals with spontaneously occurring tumors. J Transl Med. 12:2252014. View Article : Google Scholar : PubMed/NCBI

6 

Holm E, Hagmüller E, Staedt U, Schlickeiser G, Günther HJ, Leweling H, Tokus M and Kollmar HB: Substrate balances across colonic carcinomas in humans. Cancer Res. 55:1373–1378. 1995.PubMed/NCBI

7 

Vaupel P, Kallinowski F and Okunieff P: Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: A review. Cancer Res. 49:6449–6465. 1989.PubMed/NCBI

8 

Stubbs M, McSheehy PM and Griffiths JR: Causes and consequences of acidic pH in tumors: A magnetic resonance study. Adv Enzyme Regul. 39:13–30. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Stubbs M, Rodrigues L, Howe FA, Wang J, Jeong KS, Veech RL and Griffiths JR: Metabolic consequences of a reversed pH gradient in rat tumors. Cancer Res. 54:4011–4016. 1994.PubMed/NCBI

10 

Fais S: Proton pump inhibitor-induced tumour cell death by inhibition of a detoxification mechanism. J Intern Med. 267:515–525. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Sachs G, Shin JM and Howden CW: Review article: The clinical pharmacology of proton pump inhibitors. Aliment Pharmacol Ther. 23:(Suppl 2). 2–8. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Gu M, Zhang Y, Zhou X, Ma H, Yao H and Ji F: Rabeprazole exhibits antiproliferative effects on human gastric cancer cell lines. Oncol Lett. 8:1739–1744. 2014.PubMed/NCBI

13 

Zhang B, Yang Y, Shi X, Liao W, Chen M, Cheng AS, Yan H, Fang C, Zhang S, Xu G, et al: Proton pump inhibitor pantoprazole abrogates adriamycin-resistant gastric cancer cell invasiveness via suppression of Akt/GSK-β/β-catenin signaling and epithelial-mesenchymal transition. Cancer Lett 356 (2 Pt B). 704–712. 2015. View Article : Google Scholar

14 

Bitarte N, Bandres E, Boni V, Zarate R, Rodriguez J, Gonzalez-Huarriz M, Lopez I, Sola J Javier, Alonso MM, Fortes P, et al: MicroRNA-451 is involved in the self-renewal, tumorigenicity, and chemoresistance of colorectal cancer stem cells. Stem Cells. 29:1661–1671. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Levina V, Marrangoni A, Wang T, Parikh S, Su Y, Herberman R, Lokshin A and Gorelik E: Elimination of human lung cancer stem cells through targeting of the stem cell factor-c-kit autocrine signaling loop. Cancer Res. 70:338–346. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Van den Broeck A, Vankelecom H, Van Delm W, Gremeaux L, Wouters J, Allemeersch J, Govaere O, Roskams T and Topal B: Human pancreatic cancer contains a side population expressing cancer stem cell-associated and prognostic genes. PLoS One. 8:e739682013. View Article : Google Scholar : PubMed/NCBI

17 

Yamashita T, Honda M, Nakamoto Y, Baba M, Nio K, Hara Y, Zeng SS, Hayashi T, Kondo M, Takatori H, et al: Discrete nature of EpCAM+ and CD90+ cancer stem cells in human hepatocellular carcinoma. Hepatology. 57:1484–1497. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Beck B, Driessens G, Goossens S, Youssef KK, Kuchnio A, Caauwe A, Sotiropoulou PA, Loges S, Lapouge G, Candi A, et al: A vascular niche and a VEGF-Nrp1 loop regulate the initiation and stemness of skin tumours. Nature. 478:399–403. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Bu P, Chen KY, Chen JH, Wang L, Walters J, Shin YJ, Goerger JP, Sun J, Witherspoon M, Rakhilin N, et al: A microRNA miR-34a-regulated bimodal switch targets Notch in colon cancer stem cells. Cell Stem Cell. 12:602–615. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Chaffer CL, Marjanovic ND, Lee T, Bell G, Kleer CG, Reinhardt F, D'Alessio AC, Young RA and Weinberg RA: Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell. 154:61–74. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Scaffidi P and Misteli T: In vitro generation of human cells with cancer stem cell properties. Nat Cell Biol. 13:1051–1061. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA and Lander ES: Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell. 138:645–659. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Fagoonee S, Li H, Zhang H, Altruda F and Pellicano R: Gastric cancer as a stem-cell disease: Data and hypotheses. Panminerva Med. 56:289–300. 2014.PubMed/NCBI

24 

Gao G, Sun Z, Wenyong L, Dongxia Y, Zhao R and Zhang X: A preliminary study of side population cells in human gastric cancer cell line HGC-27. Ann Transplant. 20:147–153. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Xu ZY, Tang JN, Xie HX, Du YA, Huang L, Yu PF and Cheng XD: 5-Fluorouracil chemotherapy of gastric cancer generates residual cells with properties of cancer stem cells. Int J Biol Sci. 11:284–294. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Yang Z, Guo L, Liu D, Sun L, Chen H, Deng Q, Liu Y, Yu M, Ma Y, Guo N, et al: Acquisition of resistance to trastuzumab in gastric cancer cells is associated with activation of IL-6/STAT3/Jagged-1/Notch positive feedback loop. Oncotarget. 6:5072–5087. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Arocho A, Chen B, Ladanyi M and Pan Q: Validation of the 2-DeltaDeltaCt calculation as an alternate method of data analysis for quantitative PCR of BCR-ABL P210 transcripts. Diagn Mol Pathol. 15:56–61. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Yeo M, Kim DK, Kim YB, Oh TY, Lee JE, Cho SW, Kim HC and Hahm KB: Selective induction of apoptosis with proton pump inhibitor in gastric cancer cells. Clin Cancer Res. 10:8687–8696. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Lobo NA, Shimono Y, Qian D and Clarke MF: The biology of cancer stem cells. Annu Rev Cell Dev Biol. 23:675–699. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Huang S, Chen M, Ding X, Zhang X and Zou X: Proton pump inhibitor selectively suppresses proliferation and restores the chemosensitivity of gastric cancer cells by inhibiting STAT3 signaling pathway. Int Immunopharmacol. 17:585–592. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Lichner Z, Saleh C, Subramaniam V, Seivwright A, Prud'homme GJ and Yousef GM: miR-17 inhibition enhances the formation of kidney cancer spheres with stem cell/tumor initiating cell properties. Oncotarget. 6:5567–5581. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Prud'homme GJ: Cancer stem cells and novel targets for antitumor strategies. Curr Pharm Des. 18:2838–2849. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Reya T, Morrison SJ, Clarke MF and Weissman IL: Stem cells, cancer, and cancer stem cells. Nature. 414:105–111. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Yeo M, Kim DK, Park HJ, Cho SW, Cheong JY and Lee KJ: Blockage of intracellular proton extrusion with proton extrusions with proton pump inhibitor induces apoptosis in gastric cancer. Cancer Sci. 99:1852008.

Related Articles

Journal Cover

December-2016
Volume 36 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
Feng S, Zheng Z, Feng L, Yang L, Chen Z, Lin Y, Gao Y and Chen Y: Proton pump inhibitor pantoprazole inhibits the proliferation, self‑renewal and chemoresistance of gastric cancer stem cells via the EMT/β‑catenin pathways. Oncol Rep 36: 3207-3214, 2016
APA
Feng, S., Zheng, Z., Feng, L., Yang, L., Chen, Z., Lin, Y. ... Chen, Y. (2016). Proton pump inhibitor pantoprazole inhibits the proliferation, self‑renewal and chemoresistance of gastric cancer stem cells via the EMT/β‑catenin pathways. Oncology Reports, 36, 3207-3214. https://doi.org/10.3892/or.2016.5154
MLA
Feng, S., Zheng, Z., Feng, L., Yang, L., Chen, Z., Lin, Y., Gao, Y., Chen, Y."Proton pump inhibitor pantoprazole inhibits the proliferation, self‑renewal and chemoresistance of gastric cancer stem cells via the EMT/β‑catenin pathways". Oncology Reports 36.6 (2016): 3207-3214.
Chicago
Feng, S., Zheng, Z., Feng, L., Yang, L., Chen, Z., Lin, Y., Gao, Y., Chen, Y."Proton pump inhibitor pantoprazole inhibits the proliferation, self‑renewal and chemoresistance of gastric cancer stem cells via the EMT/β‑catenin pathways". Oncology Reports 36, no. 6 (2016): 3207-3214. https://doi.org/10.3892/or.2016.5154