Open Access

Characterization of sonic hedgehog inhibition in gastric carcinoma cells

  • Authors:
    • Ruxue Bai
    • Hongchuan Zhao
    • Xiang Zhang
    • Shiyu Du
  • View Affiliations

  • Published online on: March 11, 2014     https://doi.org/10.3892/ol.2014.1964
  • Pages: 1381-1384
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Aberrant activation of the sonic hedgehog (Shh) signaling pathway plays an important role in gastric cancer. The exact mechanisms defining how the Shh pathway promotes tumorigenesis or regulates its downstream targets remains elusive. In the present study, the effects of inhibiting the Shh signaling pathway in gastric cancer AGS cells was examined. It was identified that the Shh antagonist, cyclopamine, inhibited cancer proliferation, migration and invasion in a dose‑ and time‑dependent manner. Additionally, it was revealed that several key targets that are activated by the Shh signaling pathway, Gli1 and CXCR4, were downregulated at an RNA and protein level by cyclopamine. The results from the present study may be of benefit in facilitating the development of novel therapeutic strategies to treat gastric cancer in human patients.

Introduction

The hedgehog signaling pathway is critical for it’s role in normal cell differentiation and embryonic development, as well as in the pathological processes that drive cancer formation (13). The ligands of sonic hedgehog (Shh) bind to the transmembrane receptor, Patched (ptch) 1 and 2, to relieve the suppression of the transmembrane protein, Smoothened (Smo). This subsequently triggers the nuclear translocation of various transcription factors to activate downstream target genes (2,4). In various types of cancer, including ovarian (5), lung (6,7), breast (8), prostate (9), endometrial (10), skin (11) and gastrointestinal (1214), aberrant activation of Smo genes and loss of function mutations in the ptch gene relieve the suppression of the Smo protein and trigger full-length Gli1 translocation into the nucleus, prompting excessive activation of downstream genes, including c-myc and vascular endothelial growth factor (VEGF). It has also been demonstrated that inhibition of the Shh pathway by a Smo inhibitor, such as cyclopamine, slows or prevents the growth of tumor tissues (1517).

In the case of gastric cancer cells, excessive Shh signaling activities are well known to affect cancer cell proliferation, migration and invasion, and overexpression of Shh was identified in intestinal metaplasia and stomach adenomas (18). In in vitro studies, the Shh pathway and downstream genes/proteins are highly involved in the proliferation and migration of various gastric cancer cell lines, including MKN1/7/45/74, MKN45 and AGS cells (19,20). However, the exact mechanisms defining how the Shh pathway regulates gastric tumorigenesis remains elusive.

In the present study, via the application of cyclopamine, the Shh signaling pathway was inhibited in the human gastric cancer cell line, AGS, and the effect on cell proliferation, migration and invasion was evaluated. Furthermore, it was demonstrated that the molecular and cellular expression of key Shh signaling pathway-associated factors, Gli1 and CXCR4, were markedly downregulated by cyclopamine in AGS cells.

Materials and methods

Cell culture and treatment

Human gastric cancer cell line AGS was obtained from American Type Culture Collection (ATCC CRL-1739) and were maintained in RPMI-1640 medium supplemented with 10% fetal bovine serum (Invitrogen Life Technologies, Carlsbad, CA, USA) and 100 U/ml penicillin/streptomycin. The cells were cultured either with cyclopamine (5–100 μM; Calbiochem, La Jolla, CA, USA) or without cyclopamine for 24, 48 or 72 h.

Cell proliferation assay

Cells were plated at a concentration of 2.5×104 cells/ml of culture medium in 96-well plates for 24 and 72 h. Following the defined culture periods, an MTT assay (Sigma, St. Louis, MO, USA) was applied according to the manufacturer’s instructions to calculate the volume of viable cells (21).

Apoptosis assay

Following in vitro culture for 24 h, the gastric cancer cells, a total amount of 1×106, were collected in a binding buffer (10 mM HEPES/NaOH, 140 mM NaCl, 2.5 mM CaCl2) after washing with phosphate-buffered saline (PBS; 3×10 min). Fluorescence-activated cell sorting analysis for apoptosis was conducted using an Annexin V-FITC/7-AAD kit according to the manufacturer’s instructions (Beckman Coulter, Miami, FL, USA). The mixture was incubated for 10 min in a dark room at room temperature and the stained cells were immediately analyzed using a flow cytometer (Cell Lab Quanta SC; Beckman Coulter) to determine the percentage of apoptotic cells.

Invasion assay

Cancer cell migration/invasion was performed by a quantitative cell migration assay (ECM500; Chemicon, Temecula, CA, USA) according to the manufacturer’s instructions. Warm Knockout DMEM (Sigma) in the amount of 200 μl was applied to the extracellular matrix (ECM) layer to hydrate for 2 h at room temperature. AGS cells were then dislodged by trypsinization (0.25% trypsin; Sigma) and dispersed into a homogeneous single-cell suspension at the concentration of 5×105 cells/ml, followed by washing and resuspension in Knockout DMEM. Then, cell suspension of 200 μl was allowed to adhere to the surface at 37°C for 60 min. The migration mediums containing cyclopamine were then put into the bottom chamber. Following 24 h of incubation at 37°C, 5% CO2 in air, the cells in the upper chamber were stained for 20 min, and dissolved in 10% acetic acid and the optical density (OD) was read at 560 nm on a standard reader.

Quantitative polymerase chain reaction (qPCR)

A TRIzol reagent (Roche) was used to isolate total RNA from 5×106 cells according to the manufacturer’s instructions. First-strand cDNA synthesis and amplification was conducted using an MBI Revert Aid First Strand cDNA Synthesis kit (MBI Fermentas, Amherst NY, USA). The qPCR was performed using an iQ5 Multicolor Real-Time PCR Detection system (Bio-Rad, Hercules, CA, USA). The cycle threshold values were read from the ABI 7000 software. The primers were: Forward, 5′-TCCTTTGGGGTCCAGCCTTG-3′ and reverse, 5′-ATGCCTGTGGAGTTGGGGCT-3′ for Gli1; forward, 5′-TCAGTCTGGACCGCTACCTG-3′ and reverse, 5′-CCACCCACAAGTCATTGGGG-3′ for CXCR4; and forward, 5′-AGGTCGGAGTCAACGGATTTG-3′ and reverse, 5′-GTGATGGCATGGACTGTGGT-3′ for GAPDH.

Western blot analysis

RIPA buffer (50 mM Tris, 150 mM NaCl, 1% Triton X-100, 0.1% sodium dodecyl sulfate and 1% Na-deoxycholate; pH 7.4) supplemented with protease inhibitor was used to collect the cell suspension for the western blot analysis and a Bio-Rad protein assay (Bio-Rad) was used to calculate the total protein concentrations. Briefly, the protein lysates were resolved by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred onto nitrocellulose membranes (Hybond™-P; Amersham Biosciences, Piscataway, NJ, USA). The membrane was blocked using 0.2% Tween-20 and 5% non-fat dry milk in PBS. The lysates were incubated with a primary antibodies: GLI-1 rabbit polyclonal anti human IgG (H-300, Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and CXCR-4 rabbit polyclonal IgG anti-human (H-118, Santa Cruz Biotechnology, Inc.) and a horseradish peroxidase-labeled rabbit IgG secondary antibody (Santa Cruz Biotechnology, Inc.) and detected using X-ray film.

Statistical analysis

Data were calculated in triplicate and expressed as the mean ± standard error of the mean. Comparisons were made using either student’s t-test or one-way analysis of variance post hoc tests. P<0.05 was considered to indicate a statistically significant result.

Results

Inhibition of gastric cancer cell proliferation by cyclopamine

AGS cells were cultured with or without cyclopamine for 24, 48 and 72 h, and the effect of cyclopamine on cell proliferation was measured (Fig. 1). The results demonstrated that when AGS cells were treated with 5 or 10 μM of cyclopamine for 24, 48 or 72 h, the proliferation densities were unaffected, as compared with the control conditions (P>0.05). This indicated that the application of cyclopamine at lower concentrations did not alter the cell proliferation rate. However, while AGS cells that were treated with 50 or 100 μM cyclopamine for 48 or 72 h, respectively, cell proliferation was significantly inhibited, indicating that a higher concentration of cyclopamine inhibited the growth of AGS cells in a dose-dependent manner (P<0.05).

Induction of apoptosis in gastric cancer cells by cyclopamine

Secondly, the effects of cyclopamine on the AGS cells were examined. The cells were either untreated (control) or treated with cyclopamine (50 or 100 μM) for 24 or 48 h, followed by annexin V staining. The results demonstrated that high concentrations of cyclopamine (50 or 100 μM) induced significant apoptosis in AGS cells (Table I).

Table I

Cyclopamine induces apoptosis in gastric cancer cells.

Table I

Cyclopamine induces apoptosis in gastric cancer cells.

ParameterControl50 μM100 μM
Rate of apoptosis, 24 h1.52±0.5115.25±2.11a22.55±1.94a
Rate of apoptosis, 48 h3.15±0.6324.32±2.37a30.12±2.33a

a P<0.05, as compared with the control.

Inhibition of cell invasion in gastric cancer cells by cyclopamine

A characteristic feature of gastric cancer cells is their aggressive ability to filtrate and invade a reconstituted basement membrane. The effect of cyclopamine on the cellular invasion of human gastric cancer cells was assessed in the present study. The cancer cells were either untreated (control) or treated with cyclopamine at concentrations of 10, 50 and 100 μM, and maintained in the culture medium for 24 h (Fig. 2). When treated with 10 μM of cyclopamine, AGS cells demonstrated a similar rate of invasion, as compared with that of the control condition (P>0.05). However, with higher concentrations of cyclopamine (50 and 100 μM), the baseline invasions were significantly inhibited. This response was dose-dependent as the greater the concentration of cyclopamine was, the higher the degree of inhibition it induced on cancer cell migration (P<0.05).

Downregulation of Shh-associated factors by cyclopamine in gastric cancer cells

The effects of cyclopamine on gene regulation in AGS cells are demonstrated in Fig. 3. AGS cells were treated with 10, 50 and 100 μM cyclopamine for 24 h. This identified that the higher concentrations of cyclopamine (50 and 100 μM) markedly downregulated the gene expression of Gli1 and CXCR4 in the gastric cancer cells.

Cyclopamine downregulated Shh-associated proteins in AGS cells

The effects of cyclopamine on Shh-related protein expression in AGS cells are presented in Fig. 4. The results were consistent with the gene expression results, as higher concentrations of cyclopamine (50 and 100 μM) downregulated the protein expression of Gli1 and CXCR4 in the gastric cancer cells.

Discussion

The Shh signaling pathway is important in cell differentiation and maturation (13,22). However, aberrant activation of the Shh pathway results in the proliferation of various cancer cell types, including lung, pancreatic and gastric (5,8,2325).

While the mechanisms of the Shh signaling pathway in promoting gastric tumor formation remain elusive, and the downstream targeting genes continue to be largely unknown, recent studies have indicated that various key factors, including Gil1 and CXCR4, are closely associated with these pathological processes. These studies identified that the chemokine receptor, CXCR4 and its cognate ligand, CXCL12 were expressed in cancerous tissues and possibly modulated the migration and invasion of tumors in prostate, endometrial and breast cancer (2629). The in vivo and in vitro studies have identified that CXCR4 was expressed in gastric carcinoma and gastric cancer cell lines, and correlated with the late developmental stages of lymph node cancer (30).

In the present study, it was demonstrated that, following the inhibition of the Shh pathway through the application of cyclopamine, the proliferation rates and migration capacities in gastric cancer cells were significantly reduced in response to high concentrations of the compound. In addition, it was revealed that the gene and protein expression levels of Gli1 and CXCR4 were consistently downregulated in the gastric cancer cells when high concentrations of cyclopamine were applied. These results were consistent with previous studies that demonstrated that Gli1 and CXCR4 contributed to tumorigenesis in types of cancer other than gastric (23,31,32). In conclusion, the results of the present study provide invaluable insights into the mechanisms of Shh signaling for the regulation of gastric cancer cell growth in vitro and these data may ultimately facilitate the development of novel therapeutic targets for the treatment gastric of cancer in human patients.

References

1 

Hooper JE and Scott MP: Communicating with Hedgehogs. Nat Rev Mol Cell Biol. 6:306–317. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Ingham PW and McMahon AP: Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 15:3059–3087. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Pasca di Magliano M and Hebrok M: Hedgehog signalling in cancer formation and maintenance. Nat Rev Cancer. 3:903–911. 2003.PubMed/NCBI

4 

Bale AE and Yu KP: The hedgehog pathway and basal cell carcinomas. Hum Mol Genet. 10:757–762. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Liao X, Siu MK, Au CW, et al: Aberrant activation of hedgehog signaling pathway in ovarian cancers: effect on prognosis, cell invasion and differentiation. Carcinogenesis. 30:131–140. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Watkins DN, Berman DM, Burkholder SG, Wang B, Beachy PA and Baylin SB: Hedgehog signalling within airway epithelial progenitors and in small-cell lung cancer. Nature. 422:313–317. 2003. View Article : Google Scholar

7 

Gialmanidis IP, Bravou V, Amanetopoulou SG, Varakis J, Kourea H and Papadaki H: Overexpression of hedgehog pathway molecules and FOXM1 in non-small cell lung carcinomas. Lung Cancer. 66:64–74. 2009. View Article : Google Scholar : PubMed/NCBI

8 

ten Haaf A, Bektas N, von Serenyi S, et al: Expression of the glioma-associated oncogene homolog (GLI) 1 in human breast cancer is associated with unfavourable overall survival. BMC Cancer. 9:2982009.PubMed/NCBI

9 

Karhadkar SS, Bova GS, Abdallah N, et al: Hedgehog signalling in prostate regeneration, neoplasia and metastasis. Nature. 431:707–712. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Feng YZ, Shiozawa T, Miyamoto T, et al: Overexpression of hedgehog signaling molecules and its involvement in the proliferation of endometrial carcinoma cells. Clin Cancer Res. 13:1389–1398. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Daya-Grosjean L and Couvé-Privat S: Sonic hedgehog signaling in basal cell carcinomas. Cancer Lett. 225:181–192. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Berman DM, Karhadkar SS, Maitra A, et al: Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumours. Nature. 425:846–851. 2003. View Article : Google Scholar

13 

Mori Y, Okumura T, Tsunoda S, Sakai Y and Shimada Y: Gli-1 expression is associated with lymph node metastasis and tumor progression in esophageal squamous cell carcinoma. Oncology. 70:378–389. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Qualtrough D, Buda A, Gaffield W, Williams AC and Paraskeva C: Hedgehog signalling in colorectal tumour cells: induction of apoptosis with cyclopamine treatment. Int J Cancer. 110:831–837. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Chen JK, Taipale J, Cooper MK and Beachy PA: Inhibition of Hedgehog signaling by direct binding of cyclopamine to Smoothened. Genes Dev. 16:2743–2748. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Chen JK, Taipale J, Young KE, Maiti T and Beachy PA: Small molecule modulation of Smoothened activity. Proc Natl Acad Sci USA. 99:14071–14076. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Lin TL and Matsui W: Hedgehog pathway as a drug target: Smoothened inhibitors in development. Onco Targets Ther. 5:47–58. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Lee SY, Han HS, Lee KY, et al: Sonic hedgehog expression in gastric cancer and gastric adenoma. Oncol Rep. 17:1051–1055. 2007.PubMed/NCBI

19 

Ohta M, Tateishi K, Kanai F, et al: p53-Independent negative regulation of p21/cyclin-dependent kinase-interacting protein 1 by the sonic hedgehog-glioma-associated oncogene 1 pathway in gastric carcinoma cells. Cancer Res. 65:10822–10829. 2005. View Article : Google Scholar

20 

Fukaya M, Isohata N, Ohta H, et al: Hedgehog signal activation in gastric pit cell and in diffuse-type gastric cancer. Gastroenterology. 131:14–29. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Fan XG, Kelleher D, Fan XJ, Xia HX and Keeling PW: Helicobacter pylori increases proliferation of gastric epithelial cells. Gut. 38:19–22. 1996. View Article : Google Scholar : PubMed/NCBI

22 

McMahon AP, Ingham PW and Tabin CJ: Developmental roles and clinical significance of hedgehog signaling. Curr Top Dev Biol. 53:1–114. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Yoo YA, Kang MH, Kim JS and Oh SC: Sonic hedgehog signaling promotes motility and invasiveness of gastric cancer cells through TGF-beta-mediated activation of the ALK5-Smad 3 pathway. Carcinogenesis. 29:480–490. 2008. View Article : Google Scholar

24 

Nagai S, Nakamura M, Yanai K, et al: Gli1 contributes to the invasiveness of pancreatic cancer through matrix metalloproteinase-9 activation. Cancer Sci. 99:1377–1384. 2008. View Article : Google Scholar

25 

Feldmann G, Dhara S, Fendrich V, et al: Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res. 67:2187–2196. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Raman D, Baugher PJ, Thu YM and Richmond A: Role of chemokines in tumor growth. Cancer Lett. 256:137–165. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Salvucci O, Bouchard A, Baccarelli A, et al: The role of CXCR4 receptor expression in breast cancer: a large tissue microarray study. Breast Cancer Res Treat. 97:275–283. 2006. View Article : Google Scholar

28 

Kodama J, Hasengaowa, Seki N, Kusumoto T and Hiramatsu Y: Expression of the CXCR4 and CCR7 chemokine receptors in human endometrial cancer. Eur J Gynaecol Oncol. 28:370–375. 2007.PubMed/NCBI

29 

Engl T, Relja B, Marian D, et al: CXCR4 chemokine receptor mediates prostate tumor cell adhesion through alpha5 and beta3 integrins. Neoplasia. 8:290–301. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Lee HJ, Kim SW, Kim HY, et al: Chemokine receptor CXCR4 expression, function, and clinical implications in gastric cancer. Int J Oncol. 34:473–480. 2009.PubMed/NCBI

31 

Yoon JW, Gilbertson R, Iannaccone S, Iannaccone P and Walterhouse D: Defining a role for Sonic hedgehog pathway activation in desmoplastic medulloblastoma by identifying GLI1 target genes. Int J Cancer. 124:109–119. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Katoh M: Integrative genomic analyses of CXCR4: transcriptional regulation of CXCR4 based on TGFbeta, Nodal, Activin signaling and POU5F1, FOXA2, FOXC2, FOXH1, SOX17, and GFI1 transcription factors. Int J Oncol. 36:415–420. 2010.

Related Articles

Journal Cover

May-2014
Volume 7 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
Bai R, Zhao H, Zhang X and Du S: Characterization of sonic hedgehog inhibition in gastric carcinoma cells. Oncol Lett 7: 1381-1384, 2014
APA
Bai, R., Zhao, H., Zhang, X., & Du, S. (2014). Characterization of sonic hedgehog inhibition in gastric carcinoma cells. Oncology Letters, 7, 1381-1384. https://doi.org/10.3892/ol.2014.1964
MLA
Bai, R., Zhao, H., Zhang, X., Du, S."Characterization of sonic hedgehog inhibition in gastric carcinoma cells". Oncology Letters 7.5 (2014): 1381-1384.
Chicago
Bai, R., Zhao, H., Zhang, X., Du, S."Characterization of sonic hedgehog inhibition in gastric carcinoma cells". Oncology Letters 7, no. 5 (2014): 1381-1384. https://doi.org/10.3892/ol.2014.1964