Possible regulatory role of Snail in NF-κB-mediated changes in E-cadherin in gastric cancer

  • Authors:
    • Zenan Hu
    • Xiaojun Liu
    • Zhifeng Tang
    • Yongning Zhou
    • Liang Qiao
  • View Affiliations

  • Published online on: December 19, 2012     https://doi.org/10.3892/or.2012.2200
  • Pages: 993-1000
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In the present study, we aimed to investigate the involvement of Snail in NF-κB-mediated changes of E-cadherin in gastric cancer. A total of 189 human gastric cancer tissues, and 32 normal gastric mucosal tissues were used to determine the expression levels of NF-κB, E-cadherin and Snail by immunohistochemistry. The correlation between the expression levels and patient clinicopathological data was analyzed. Human gastric cancer cell line SGC7901 was treated with the NF-κB inhibitor PDTC, and the expression levels of E-cadherin and Snail were investigated by qPCR and western blot. NF-κB, E-cadherin and Snail were all detected in normal gastric mucosa and cancer tissues of various differentiation statuses. However, the expression patterns of each protein were different. Strong expression of E-cadherin was detected in normal gastric mucosa, whereas its expression gradually declined in gastric cancer tissues, with weak expression observed in poorly differentiated gastric cancer tissues. In contrast, weak NF-κB and Snail expressions were present in normal gastric mucosa, while their expression levels gradually increased in gastric cancer tissues, with the strongest expression detected in poorly differentiated gastric cancers. The expression of E-cadherin was inversely correlated with that of Snail and NF-κB in the tissues tested. Blockade of NF-κB using its inhibitor PDTC led to a time-dependent reduction in Snail but a time-dependent increase in E-cadherin in SGC7901 cells. These results suggest that in human gastric cancer, loss of E-cadherin may be mediated through NF-κB-induced Snail upregulation. Further studies may reveal whether targeting the NF-κB-Snail-E-cadherin axis could be a useful approach for combating gastric cancer.

Introduction

Gastric cancer is a major public health issue worldwide particularly in China. According to cancer statistics published in 2011, gastric cancer is the fourth most frequently diagnosed cancer and the third most common cause of cancer-related mortality in men, whereas in women it is the fifth most common malignancy in regards to incidence and mortality rate (1,2). The highest incidence rates of gastric cancer are in Eastern Asia, Eastern Europe and South America (1,2). In China, gastric cancer is the third most common malignancy and the leading cause of cancer-related death (3,4). Lack of effective treatment options for advanced gastric cancer is largely due to a poor understanding of the molecular mechanisms involved in the development of gastric cancer.

Nuclear factor-κB (NF-κB) is a ubiquitously expressed family of Rel-related transcription factors (5). Abnormal activation of NF-κB reduces cell sensitivity to apoptotic stimuli and therefore facilitates the survival of transformed cells (6). NF-κB is involved in the control of cell growth and oncogenesis. Constitutive activation of NF-κB in cancer cells is partially responsible for the observed resistance to chemotherapy and radiotherapy (7). As a ubiquitous transcription factor, NF-κB regulates the expression and function of numerous target genes, among which and of most relevance to cancer development is E-cadherin.

E-cadherin is a major cell-cell adhesion molecule that plays a significant role in the establishment and maintenance of cell-cell interactions and tissue architecture (810). A negative correlation between NF-κB and E-cadherin in gastric cancer cells has recently been reported (11). It was recently shown that connective tissue growth factor (CTGF) downregulated the expression of E-cadherin through activation of NF-κB (11). Loss of E-cadherin expression is associated with enhanced tumor progression, increased invasive and metastatic potential of cancer cells and a poor overall prognosis in patients with gastric cancer and other malignancies (1215). However, as gastric cancer is a multifactorial disease (16), loss of E-cadherin alone cannot explain the increased malignant tendency of gastric cancer cells (17). Interaction between E-cadherin and other genes could well be involved in the development of gastric cancer and its malignant phenotype.

We supposed that Snail may be a critical factor in mediating the regulatory role of NF-κB on its target genes, which has not been reported in the literature. Snail is a member of the Snail superfamily of zinc finger transcription factors (18). It plays an important role in embryonic development, neural differentiation, cell division and survival (19,20). Overexpression of Snail mRNA was able to downregulate the expression of E-cadherin in diffuse-type gastric carcinoma (21,22). However, it is not clear whether Snail is a critical transcription factor for the regulatory role of NF-κB regarding its target genes.

This study aimed to evaluate whether NF-κB-mediated changes in E-cadherin are regulated through Snail.

Materials and methods

Donor blocks and patient information

Paraffin-embedded blocks of gastric tissues (previously fixed in 10% formaldehyde) were obtained from 189 patients with gastric cancer who underwent surgical operations at the Wuwei Tumor Hospital, Gansu Province, China. The diagnosis of gastric adenocarcinoma was based on the World Health Organization (WHO) diagnostic criteria, and was confirmed by two independent pathologists. Based on the WHO Classification of Tumors of the Digestive System (23), there were 100 cases of poorly differentiated gastric adenocarcinoma, 44 cases of moderately differentiated gastric adenocarcinoma, and 45 cases of well-differentiated gastric adenocarcinoma. The patient study population had a mean age of 55 (range, 30–73) years at the time of operation, with an overall male to female ratio of 3.3:1. None of the patients had received any chemotherapy and/or radiotherapy prior to surgery. The detailed patient characteristics are summarized in Table I. Paraffin-embedded blocks of normal gastric mucosal tissues (n=32) were obtained from healthy subjects who underwent gastroscopy in the same hospital for other non-malignant gastric conditions. Written consent from all patients was obtained prior to the study. The study was approved by the Institutional Human Ethics Committee of the First Clinical School of Lanzhou University.

Table I

Clinicopathological features of the 189 patients with gastric cancer.

Table I

Clinicopathological features of the 189 patients with gastric cancer.

CharacteristicsNo. of cases%
Gender
 Female4423.3
 Male14576.7
Age (years)
 <505127
 ≥5013873
Tumor size (cm)
 <57037
 ≥511963
Lymph node metastasis
 No7338.6
 Yes11661.4
Tumor differentiation status
 Well/moderate8947.1
 Poor10052.9
Depth of tumor invasion
 Without serosal invasion4624.3
 Serosal invasion14375.7
Lauren classification
 Intestinal type9751.3
 Diffuse type8548.7
Tissue microarray (TMA) construction

The collected paraffin blocks were used as donor blocks to make eight TMA recipient blocks. In each donor block, morphologically representative areas were chosen and marked on their respective H&E slides. A tissue core of 0.6 mm in diameter from each donor block was taken using a cylindrical tissue puncher (Beecher, Beecher Instruments, Silver Spring, MD, USA) and transferred into the hole on the recipient paraffin block. The distance between each recipient hole was kept constant at 1 mm. Duplicate tissue cores from each donor tissue were positioned side by side. The detailed matrix plan for the arrangement of the constructed TMA was recorded for correct tissue identification.

Immunohistochemistry assays

The above-constructed TMA blocks were cut into sections of 4-μm thickness, dewaxed in xylene and rehydrated in graded alcohols. The slides were boiled for 30 min in citrate buffer (10 mM; pH 6.0) in a microwave oven at 250–300 W and then cooled to room temperature. Before immunohistochemical staining, the slides were incubated with 3% H2O2 in PBS for 10 min to quench the endogenous peroxidase activity, followed by incubation with 3% BSA for 15 min to block the non-specific binding of the antibody.

For immunohistochemical staining, the slides were incubated for 1 h at 37°C with primary antibody against E-cadherin (monoclonal, dilution 1:250, Abcam, USA), NF-κB p65 (monoclonal, dilution 1:200, Abcam), and Snail (polyclonal, dilution 1:200, Abcam). The slides were then washed with PBS for three times, incubated with biotin-conjugated secondary antibody (1:150, Abcam) for 40 min at 37°C, washed with PBS, and then incubated with streptavidin-horseradish peroxidase (SHRP) (Thermo Fisher Scientific, USA) for 40 min at room temperature. DAB (2,3-diaminobenzidine tetrahydrochloride) (Beijing Zhongshan Golden Bridge Biotechnology Co., Ltd., China) was used to develop the peroxidase reaction, and the slides were counterstained with hematoxylin. The experimental validity was confirmed by using negative controls in which the primary antibody was replaced by 5% BSA. The slides were reviewed independently by two pathologists, and the staining for each protein was scored according to the criteria established in Table II and as previously reported (24). Representative areas were photographed for data presentation.

Table II

Scoring criteria for immunohistochemistry.

Table II

Scoring criteria for immunohistochemistry.

CriteriaScore
Staining positivity
 Positive in <5% of the cells0
 Positive in 5–25% of the cells1
 Positive in 26–50% of the cells2
 Positive in >50% of the cells3
Staining intensity
 Negative (no staining)0
 Weak (light yellow)1
 Moderate (brown)2
 High (dark brown)3
Sum of positivity and intensity scores
 Negative0–2
 Weak positive3–4
 Strong positive5–6
Culture of gastric cancer cells and treatment with NF-κB inhibitor PDTC

SGC7901 cells (a human gastric cancer cell line; Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China) were cultured in RPMI-1640 medium (Gibco, Carlsbad, CA, USA) supplemented with 1% penicillin and streptomycin (Gibco) and 10% heat-inactivated fetal bovine serum (Hangzhou Sijiqing Biological Engineering Materials Co., Ltd., Hangzhou, China) at 37°C in a humidified atmosphere containing 5% carbon dioxide.

To block the activity of NF-κB, cells were treated with 50 μM of a chemical inhibitor of NF-κB, pyrrolidine dithiocarbamate (PDTC). This optimal dose was based on our preliminary study by sulforhodamine B (SRB) assay, which revealed that 50 μM of PDTC was able to effectively block the expression and activity of the NF-κB subunit p65 in gastric cancer cells. The SRB assay was performed as previously reported (25,26).

Quantitative real-time PCR (qPCR)

Total RNA of the treated cells was extracted using the Ze Spin Column of the Total RNA Isolation kit (Takara, Dalian, China). Total RNA (1 μg) was reverse-transcribed into cDNA using the PrimeScript™ RT Reagent kit (Takara) according to the manufacturer’s instructions. The synthesized cDNA samples were subjected to qPCR using SYBR® Premix Ex Taq™ reagent (Takara). All qPCR reactions were performed using Rotor-Gene 3000 (Corbett, Australia), with each PCR cycle consisting of denaturation for 15 sec at 95°C, annealing for 45 sec at 62°C and extension for 30 sec at 72°C. β-actin was used as the internal reference. The qPCR primers were as follows: E-cadherin (sense: 5′-TTAAACTCCTGGCCTCAAGCAATC-3′, antisense: 5′-TCCTATCTTGGGCAAAGCAACTG-3′), NF-κB/P65 (sense: 5′-TCAGTCAGCGCATCCAGACC-3′, antisense: 5′-CAGAGCCGCACAGCATTCA-3′), Snail (sense: 5′-CGC GCTCTTTCCTCGTCAG-3′, antisense: 5′-TCCCAGATGA GCATTGGCAG-3′), β-actin (sense: 5′-TGGCACCCAGCA CAATGAA-3′, antisense: 5′-CTAAGTCATAGTCCGCCTAG AAGCA-3′). For data analysis, fold induction relative to internal controls was calculated by the Δ Ct evaluation method.

Western blot assay

Total protein from the treated cells was extracted using RIPA buffer (Beyotime, Shanghai, China) supplemented with phenylmethylsulfonyl fluoride (PMSF) and protease inhibitor cocktails (Roche, Germany), and the protein concentrations were measured by a BCA protein quantitative assay kit (Applygen, Beijing, China). The cell lysates were cleared by centrifugation at 10,000 × g for 5 min at 4°C. Equal amounts of total proteins were resolved on 10% polyacrylamide gels (SDS-PAGE) and transferred to PVDF membranes, which were incubated with primary antibodies (E-cadherin, NF-κB and Snail) at a dilution of 1:1,000 overnight at 4°C. The membranes were then incubated with HRP-conjugated secondary antibody (1:10,000) for 1 h at room temperature and exposed using an enhanced chemiluminescence (ECL) detection system (Applygen) and visualized by autoradiography. β-actin was used as the internal reference.

Statistical analysis

SPSS 15.0 was used for data analysis. All values are expressed as means ± SD. The Student’s t-test was used to evaluate the difference between mean values. Immunohistochemical staining was quantitated and differences between groups were assessed by the χ2 test. A P-value of <0.05 was considered to indicate a statistically significant result.

Results

Expression pattern of E-cadherin

E-cadherin was detected in all tissues tested, including normal gastric epithelial tissues, adjacent non-cancerous gastric epithelial tissues and gastric cancer tissues. In normal gastric mucosa, strong expression of E-cadherin was present as a membranous protein, with some weak staining in the cytoplasmic compartment. In gastric cancer tissues, E-cadherin was largely expressed in cytoplasmic compartments with weak expression on the membrane. Normal gastric mucosal tissues expressed a higher level of E-cadherin (Fig. 1A) than gastric cancer tissues (Fig. 1B-D). Among the gastric cancer tissues, a higher level of E-cadherin was detected in the well/moderately differentiated cancer tissues (Fig. 1B and C) than in poorly differentiated cancer tissues (Fig. 1D). Overall, E-cadherin was detected in 22% (41/189) of gastric cancer tissues, 55.6% (30/54) of matched non-cancerous gastric tissues, and 100% (32/32) of normal gastric mucosa. By Chi-square (χ2) test, gastric cancer tissues expressed a reduced level of E-cadherin compared to the matched non-cancerous gastric tissues (χ2=22.382, P=0.000), and normal gastric mucosa (χ2=74.33, P=0.000). Of note, reduced expression of E-cadherin was observed in matched non-cancerous gastric tissues when compared with tha normal gastric mucosa (χ2=19.728, P=0.000). As shown in Table III, increased E-cadherin expression in gastric cancer tissues strongly correlated with a better differentiation status (P=0.000) and less invasion (P=0.004). By Lauren classification, higher expression level of E-cadherin was found in tumors of intestinal type than in tumors of diffuse type (P=0.002). The expression of E-cadherin did not appear to be associated with age, gender, tumor size and lymph node metastasis.

Table III

Relationship between E-cadherin expression and clinicopathological factors in 189 patients with gastric cancer.

Table III

Relationship between E-cadherin expression and clinicopathological factors in 189 patients with gastric cancer.

E-cadherin

VariablesPositiveNegativeTotalPositive rate (%)χ2P-value
Gender0.4160.519
 Female8364418.2
 Male3311214522.8
Age (years)0.6730.412
 <509425117.6
 ≥503210613823.2
Tumor size (cm)2.3820.123
 <519517027.1
 ≥5219811917.6
Lymph node metastasis2.9480.086
 No21527328.8
 Yes219511618.1
Differentiation status14.2940.000
 Well/moderate30598933.7
 Poor118910011.0
Depth of tumor invasion8.3380.004
 Without serosal invasion17294637.0
 Serosal invasion2411914316.8
Lauren classification9.7020.002
 Intestinal type30679730.9
 Diffuse type10758511.8
Expression pattern of NF-κB

NF-κB was detected in the cytoplasmic and nuclear portions of cells in normal gastric mucosa, matched non-cancerous gastric tissues and gastric cancer tissues to a various extent. Unlike E-cadherin, gastric cancer tissues (Fig. 1F-H) expressed a significantly higher level of NF-κB than non-cancerous gastric tissues (data not shown) and normal gastric mucosa (Fig. 1E). Among the gastric cancer tissues, a higher level of NF-κB was detected in poorly differentiated cancer tissues (Fig. 1H) than in well/moderately differentiated cancer tissues (Fig. 1G and F). Overall, NF-κB was detected in 75.1% (142/189) of gastric cancer tissues, 42.6% (23/54) of matched non-cancerous gastric tissues, and 15.6% (5/32) of normal gastric mucosal tissues. By χ2 test, the expression of NF-κB was significantly higher in gastric cancer tissues compared to that in the matched non-cancerous gastric tissues (χ2=20.404, P=0.000) and normal gastric mucosa (χ2=43.511, P=0.000). Matched non-cancerous gastric tissues also expressed a higher level of NF-κB than the normal gastric mucosa (χ2=6.655, P=0.010). In patients with gastric cancers, increased expression of NF-κB was found to be strongly correlated with an increased tendency for lymph node metastasis (P=0.018), deeper tumor invasion (P=0.010), poor tumor differentiation (P=0.021), and diffuse type of cancer histology (P=0.007) (Table IV). The expression of NF-κB was, however, not associated with gender, age and tumor size.

Table IV

Relationship between NF-κB expression and clinicopathological factors in 189 patients with gastric cancer.

Table IV

Relationship between NF-κB expression and clinicopathological factors in 189 patients with gastric cancer.

NF-κB

VariablesPositiveNegativeTotalPositive rate (%)χ2P-value
Gender1.4830.223
 Female30144468.2
 Male1123314577.2
Age (years)1.0340.309
 <5041105180.4
 ≥501013713873.2
Tumor size (cm)1.5670.211
 <549217070.0
 ≥5932611978.2
Lymph node metastasis5.6000.018
 No48257365.6
 Yes942211681.0
Differentiation status5.3610.021
 Well/moderate60298967.4
 Poorly821810082.0
Depth of tumor invasion6.6190.010
 Without serosal invasion28184660.9
 Serosal invasion1142914379.7
Lauren classification7.2840.007
 Intestinal type64339766.0
 Diffuse type71148583.5
Expression pattern of Snail

Snail had a similar expression pattern as NF-κB in that it was detected in the cytoplasmic and nuclear compartments of cells in normal gastric mucosa, matched non-cancerous gastric tissues, and gastric cancer tissues. Gastric cancer tissues (Fig. 1J-L) expressed a significantly higher level of Snail than non-cancerous gastric tissues (data not shown) and normal gastric mucosa (Fig. 1I). Among the gastric cancer tissues, a higher level of Snail was detected in poorly differentiated cancer tissues (Fig. 1L) than in well/moderately differentiated cancer tissues (Fig. 1J and H). Overall, Snail was detected in 75.7% (143/189) of gastric cancer tissues, 48.45% (26/54) of matched non-cancerous gastric tissues, and 18.75% (6/32) of normal gastric mucosal tissues. By χ2 test, the expression of Snail was significantly higher in gastric cancer tissues compared to that in the matched non-cancerous gastric tissues (χ2=23.67, P=0.000) and that in normal gastric mucosa (χ2=55.95, P=0.000). Matched non-cancerous gastric tissues also expressed a higher level of Snail than that in the normal gastric mucosa (χ2=7.89, P=0.010).

As shown in Table V, in patients with gastric cancer, increased expression of Snail was found to be strongly correlated with increased potential for lymph node metastasis (P=0.03), increased tumor invasion (P=0.018), poor tumor differentiation (P=0.032), and diffuse type of cancer histology (P=0.003). Similar to NF-κB, the expression of Snail was not associated with gender, age and tumor size.

Table V

Relationship between Snail expression and clinicopathological factors in the 189 patients with gastric cancer.

Table V

Relationship between Snail expression and clinicopathological factors in the 189 patients with gastric cancer.

Snail

VariablesPositiveNegativeTotalPositive rate (%)χ2P-value
Gender0.0540.816
 Female32124472.7
 Male1113414576.6
Age (years)0.6910.406
 <5040115178.4
 ≥501033513874.6
Tumor size (cm)3.0350.081
 <548227068.6
 ≥5952411979.8
Lymph node metastasis4.7080.03
 No49247367.1
 Yes942211681.0
Differentiation status4.5860.032
 Well/moderate61288968.5
 Poor821810082.0
Depth of tumor invasion5.5490.018
 Without serosal invasion29174663.0
 Serosal invasion1142914379.7
Lauren classification8.5630.003
 Intestinal type67309769.1
 Diffuse type70158582.4
Effect of NF-κB blockade on the expression of E-cadherin and Snail

The above results showed that in gastric cancer tissues, there was a close correlation between the expression of NF-κB, E-cadherin and Snail. We proposed that NF-κB may regulate the expression of E-cadherin via the transcription factor Snail. In order to examine for this, we chose gastric cancer cell line SGC7901 as a model to investigate whether modulation of NF-κB in this cell line could affect the expression of E-cadherin and Snail.

Following treatment of SGC7901 cells with 50 μM of the NF-κB inhibitor PDTC, for 0, 12, 24 and 36 h, a time-dependent reduction in NF-κB was noted at the mRNA (Fig. 2A) and protein (Fig. 2B) levels. Similarly, PDTC-induced reduction of NF-κB in SGC7901 cells was associated with a reduced expression of Snail in a time-dependent manner at both the mRNA and protein levels (Fig. 2). On the other hand, blockade of NF-κB with PDTC rendered a time-dependent increase in the expression of E-cadherin at both the mRNA and protein levels (Fig. 2).

Discussion

Gastric cancer is a multifactorial disease. Despite numerous studies, the molecular mechanisms for gastric cancer development have not yet been clarified. Our previous studies demonstrated that loss of E-cadherin contributes to the local and distant spread of gastric cancer (15,27). The expression and function of E-cadherin can be regulated by many factors such as β-catenin and NF-κB (15,28,29). Our current study suggests that in gastric cancer, increased expression and activity of NF-κB may contribute to the observed loss of E-cadherin, and this biological change may be caused through NFκB-mediated alteration in the expression of Snail.

NF-κB is a critical transcription factor involved in the regulation of many signaling pathways that are important in inflammation, the immune response and cancer development (30,31). The importance of NF-κB in the development of gastric cancer has been well-documented (3234). In the present study, we found a reverse correlation between the expression of E-cadherin and NF-κB in normal and malignant gastric tissues. High expression level of E-cadherin in normal gastric mucosa was correlated with a low level of NF-κB, whereas in malignant gastric tissues, loss of E-cadherin was correlated with an increased activity of NF-κB.

E-cadherin is a cell-cell adhesion molecule that plays an important role in the formation of cell polarity and tissue architecture (8,9). Although studies on E-cadherin-deficient mice have provided little support concerning the role of E-cadherin in the development of gastric adenocarcinoma (17), numerous studies have shown that loss of E-cadherin is closely related to increased tumor cell migration, more aggressive invasion and metastasis, and poor prognosis of gastric cancer (35,36). Additionally, E-cadherin expression negatively controls the transcriptional activity of NF-κB (29). We speculated that the inverse relationship between these two molecules may be an important mechanism in gastric cancer formation and metastasis.

The inverse correlation between E-cadherin and NF-κB was recapitulated in our in vitro study in gastric cancer cells. When NF-κB was blocked using its chemical inhibitor PDTC in SGC7901 cells (as shown by a time-dependent decrease in the NF-κB subunit p65 at the mRNA and protein levels), we observed a time-dependent increase in the expression of E-cadherin. Such an inverse correlation between NF-κB and E-cadherin may be regulated by NF-κB-regulated Snail activity, as blockade of NF-κB was also followed by a time-dependent inhibition of Snail. As blockade of NF-κB has been shown to inhibit the growth of cancer cells (3740), we believe that NF-κB-mediated cancer cell growth may be regulated through the transcription factor Snail.

Snail is an important transcription factor that has been shown to regulate many extracellular matrix genes (20,41,42). Several studies have demonstrated that Snail functions as a direct inhibitor for the transcription of E-cadherin (43,44), particularly in malignant tumors (45). In addition, Snail was recognized as an independent marker for the prognosis of patients with gastric carcinoma (46). Our study indicates that in gastric cancer cells, the regulatory effect of NF-κB on its target genes such as E-cadherin is likely mediated through Snail. To support this finding, previous studies have shown the presence of the NF-κB binding sequence on the promoter of the Snail gene (47,48).

As NF-κB plays an important role in the control of growth and survival of cancer cells, and loss of E-cadherin is closely related to the development of gastric cancer and its metastasis, our data not only provide a new mechanism of how NF-κB may regulate E-cadherin in gastric cancer, but also potentially opens a new avenue for possible therapeutic targeting. If Snail is a critical intermediating factor between NF-κB and its targets, then specific targeting of Snail may be of therapeutic benefit. Further studies using more cell lines involving specific knockdown of Snail (e.g., using siRNA) and appropriate in vivo studies are needed to generate more valuable data to confirm such an assumption.

In conclusion, our results showed that in gastric cancer, loss of E-cadherin in gastric epithelial cells may be regulated through NF-κB-mediated Snail signaling. Further studies are warranted to clarify the role of the NF-κB-Snail-E-cadherin axis in gastric cancer.

Acknowledgements

We thank Drs Zhaofeng Chen, Lina Wang, and Meikai Zhou from the First Clinical Medical School of Lanzhou University for their assistance in TMA construction and immunohistochemistry. This study was funded by the National Natural Science Funding of China (grant ID: no. 432355/041003). Dr Z. Hu’s visiting study to the Storr Liver Unit of the Westmead Millennium Institute was supported by the Robert W. Storr Bequest. Dr L. Qiao was supported by the Robert W. Storr Bequest and the Career Development and Support Fellowship Future Research Leader Grant of the NSW Cancer Institute, NSW, Australia.

Abbreviations:

NF-κB

nuclear factor-κB

PDTC

pyrrolidine dithiocarbamate

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar

2 

Siegel R, Ward E, Brawley O and Jemal A: Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 61:212–236. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Yang L, Parkin DM, Ferlay J, Li L and Chen Y: Estimates of cancer incidence in China for 2000 and projections for 2005. Cancer Epidemiol Biomarkers Prev. 14:243–250. 2005.PubMed/NCBI

4 

Yang L: Incidence and mortality of gastric cancer in China. World J Gastroenterol. 12:17–20. 2006.

5 

Baeuerle PA and Baltimore D: NF-κB: ten years after. Cell. 87:13–20. 1996.

6 

Bours V, Dejardin E, Goujon-Letawe F, Merville MP and Castronovo V: The NF-kappa B transcription factor and cancer: high expression of NF-kappa B- and I kappa B-related proteins in tumor cell lines. Biochem Pharmacol. 47:145–149. 1994. View Article : Google Scholar : PubMed/NCBI

7 

Baldwin AS: Control of oncogenesis and cancer therapy resistance by the transcription factor NF-κB. J Clin Invest. 107:241–246. 2001.

8 

Frixen UH, Behrens J, Sachs M, et al: E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. J Cell Biol. 113:173–185. 1991. View Article : Google Scholar : PubMed/NCBI

9 

Hirohashi S: Inactivation of the E-cadherin-mediated cell adhesion system in human cancers. Am J Pathol. 153:333–339. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Gabbert HE, Mueller W, Schneiders A, et al: Prognostic value of E-cadherin expression in 413 gastric carcinomas. Int J Cancer. 69:184–189. 1996. View Article : Google Scholar : PubMed/NCBI

11 

Mao Z, Ma X, Rong Y, et al: Connective tissue growth factor enhances the migration of gastric cancer through downregulation of E-cadherin via the NF-κB pathway. Cancer Sci. 102:104–110. 2011.PubMed/NCBI

12 

Ghadimi BM, Behrens J, Hoffmann I, Haensch W, Birchmeier W and Schlag PM: Immunohistological analysis of E-cadherin, alpha-, beta-and gamma-catenin expression in colorectal cancer: implications for cell adhesion and signaling. Eur J Cancer. 35:60–65. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Pignatelli M, Ansari TW, Gunter P, et al: Loss of membranous E-cadherin expression in pancreatic cancer: correlation with lymph node metastasis, high grade, and advanced stage. J Pathol. 174:243–248. 1994. View Article : Google Scholar : PubMed/NCBI

14 

Oka H, Shiozaki H, Kobayashi K, et al: Expression of E-cadherin cell adhesion molecules in human breast cancer tissues and its relationship to metastasis. Cancer Res. 53:1696–1701. 1993.PubMed/NCBI

15 

Zhou Y, Li G, Wu J, et al: Clinicopathological significance of E-cadherin, VEGF, and MMPs in gastric cancer. Tumour Biol. 31:549–558. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Kyrlagkitsis I and Karamanolis DG: Genes and gastric cancer. Hepatogastroenterology. 51:320–327. 2004.

17 

Mimata A, Fukamachi H, Eishi Y and Yuasa Y: Loss of E-cadherin in mouse gastric epithelial cells induces signet ring-like cells, a possible precursor lesion of diffuse gastric cancer. Cancer Sci. 102:942–950. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Nieto MA: The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol. 3:155–166. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Bonavida B and Baritaki S: Dual role of NO donors in the reversal of tumor cell resistance and EMT: downregulation of the NF-κB/Snail/YY1/RKIP circuitry. Nitric Oxide. 24:1–7. 2011.PubMed/NCBI

20 

Wu Y and Zhou BP: Snail: more than EMT. Cell Adh Migr. 4:199–203. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Rosivatz E, Becker I, Specht K, et al: Differential expression of the epithelial-mesenchymal transition regulators Snail, SIP1, and Twist in gastric cancer. Am J Pathol. 161:1881–1891. 2002. View Article : Google Scholar

22 

Castro Alves C, Rosivatz E, Schott C, et al: Slug is overexpressed in gastric carcinomas and may act synergistically with SIP1 and Snail in the down-regulation of E-cadherin. J Pathol. 211:507–515. 2007.PubMed/NCBI

23 

Bosman FT: WHO classification of tumours of the digestive system. World Health Organization. International Agency for Research on Cancer; 4th edition. IARC Press; Lyon: 2010

24 

Volm M, Koomagi R and Mattern J: Prognostic value of vascular endothelial growth factor and its receptor Flt-1 in squamous cell lung cancer. Int J Cancer. 74:64–68. 1997. View Article : Google Scholar : PubMed/NCBI

25 

Vichai V and Kirtikara K: Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat Protoc. 1:1112–1116. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Woolston C and Martin S: Analysis of tumor and endothelial cell viability and survival using sulforhodamine B and clonogenic assays. Methods Mol Biol. 740:45–56. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Zhou Y, Ran J, Tang C, et al: Effect of celecoxib on E-cadherin, VEGF, microvessel density and apoptosis in gastric cancer. Cancer Biol Ther. 6:269–275. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Dentice M, Luongo C, Ambrosio R, et al: Beta-catenin regulates deiodinase levels and thyroid hormone signaling in colon cancer cells. Gastroenterology. 143:1037–1047. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Solanas G, Porta-de-la-Riva M, Agustí C, et al: E-cadherin controls beta-catenin and NF-kappaB transcriptional activity in mesenchymal gene expression. J Cell Sci. 121:2224–2234. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Sebastian K, Reza F, Christoph C, et al: Time dependency and topography of hepatic NF-κB activation after hemorrhagic shock and resuscitation in mice. Shock. 38:486–492. 2012.PubMed/NCBI

31 

Bernardi FC, Felisberto F, Vuolo F, et al: Oxidative damage, inflammation, and toll-like receptor 4 pathway are increased in preeclamptic patients: a case-control study. Oxid Med Cell Longev. 2012:6364192012. View Article : Google Scholar : PubMed/NCBI

32 

Jiang Z, Wu W and Qian ML: Cellular damage and apoptosis along with changes in NF-kappa B expression were induced with contrast agent enhanced ultrasound in gastric cancer cells and hepatoma cells. Cancer Cell Int. 12:82012. View Article : Google Scholar : PubMed/NCBI

33 

Lee KH and Kim JR: Regulation of HGF-mediated cell proliferation and invasion through NF-κB, JunB, and MMP-9 cascades in stomach cancer cells. Clin Exp Metastasis. 29:263–272. 2012.PubMed/NCBI

34 

Li J, Shen L, Lu FR, et al: Plumbagin inhibits cell growth and potentiates apoptosis in human gastric cancer cells in vitro through the NF-κB signaling pathway. Acta Pharmacol Sin. 33:242–249. 2012.PubMed/NCBI

35 

Uchikado Y, Okumura H, Ishigami S, et al: Increased Slug and decreased E-cadherin expression is related to poor prognosis in patients with gastric cancer. Gastric Cancer. 14:41–49. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Maehata Y, Hirahashi M, Aishima S, et al: Significance of dysadherin and E-cadherin expression in differentiated-type gastric carcinoma with submucosal invasion. Hum Pathol. 42:558–567. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Biswas DK, Shi Q, Baily S, et al: NF-kappa B activation in human breast cancer specimens and its role in cell proliferation and apoptosis. Proc Natl Acad Sci USA. 101:10137–10142. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Dolcet X, Llobet D, Pallares J and Matias-Guiu X: NF-κB in development and progression of human cancer. Virchows Arch. 446:475–482. 2005.

39 

Han SS, Yun H, Son DJ, et al: NF-kappaB/STAT3/PI3K signaling crosstalk in iMyc E mu B lymphoma. Mol Cancer. 9:972010. View Article : Google Scholar : PubMed/NCBI

40 

Fang Y, Sun H, Zhai J, et al: Antitumor activity of NF-κB decoy oligodeoxynucleotides in a prostate cancer cell line. Asian Pac J Cancer Prev. 12:2721–2726. 2011.

41 

Dhasarathy A, Kajita M and Wade PA: The transcription factor snail mediates epithelial to mesenchymal transitions by repression of estrogen receptor-alpha. Mol Endocrinol. 21:2907–2918. 2007. View Article : Google Scholar

42 

Harder JL, Whiteman EL, Pieczynski JN, Liu CJ and Margolis B: Snail destabilizes cell surface Crumbs3a. Traffic. 13:1170–1185. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Becker KF, Rosivatz E, Blechschmidt K, Kremmer E, Sarbia M and Höfler H: Analysis of the E-cadherin repressor Snail in primary human cancers. Cells Tissues Organs. 185:204–212. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Blechschmidt K, Kremmer E, Hollweck R, et al: The E-cadherin repressor snail plays a role in tumor progression of endometrioid adenocarcinomas. Diagn Mol Pathol. 16:222–228. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Spaderna S, Schmalhofer O, Wahlbuhl M, et al: The transcriptional repressor ZEB1 promotes metastasis and loss of cell polarity in cancer. Cancer Res. 68:537–544. 2008. View Article : Google Scholar : PubMed/NCBI

46 

He H, Chen W, Wang X, et al: Snail is an independent prognostic predictor for progression and patient survival of gastric cancer. Cancer Sci. 103:1296–1303. View Article : Google Scholar

47 

Bachelder RE, Yoon SO, Franci C, de Herreros AG and Mercurio AM: Glycogen synthase kinase-3 is an endogenous inhibitor of Snail transcription: implications for the epithelial-mesenchymal transition. J Cell Biol. 168:29–33. 2005. View Article : Google Scholar

48 

Julien S, Puig I, Caretti E, et al: Activation of NF-kappaB by Akt upregulates Snail expression and induces epithelium mesenchyme transition. Oncogene. 26:7445–7456. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March 2013
Volume 29 Issue 3

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
Hu Z, Liu X, Tang Z, Zhou Y and Qiao L: Possible regulatory role of Snail in NF-κB-mediated changes in E-cadherin in gastric cancer. Oncol Rep 29: 993-1000, 2013
APA
Hu, Z., Liu, X., Tang, Z., Zhou, Y., & Qiao, L. (2013). Possible regulatory role of Snail in NF-κB-mediated changes in E-cadherin in gastric cancer. Oncology Reports, 29, 993-1000. https://doi.org/10.3892/or.2012.2200
MLA
Hu, Z., Liu, X., Tang, Z., Zhou, Y., Qiao, L."Possible regulatory role of Snail in NF-κB-mediated changes in E-cadherin in gastric cancer". Oncology Reports 29.3 (2013): 993-1000.
Chicago
Hu, Z., Liu, X., Tang, Z., Zhou, Y., Qiao, L."Possible regulatory role of Snail in NF-κB-mediated changes in E-cadherin in gastric cancer". Oncology Reports 29, no. 3 (2013): 993-1000. https://doi.org/10.3892/or.2012.2200