Herbal prescription, Danggui-Sayuk-Ga-Osuyu-Senggang-Tang, inhibits TNF-α-induced epithelial-mesenchymal transition in HCT116 colorectal cancer cells

  • Authors:
    • Kangwook Lee
    • Sung-Gook Cho
    • Youn Kyung Choi
    • Yu-Jeong Choi
    • Gyu-Ri Lee
    • Chan-Yong Jeon
    • Seong-Gyu Ko
  • View Affiliations

  • Published online on: November 7, 2017     https://doi.org/10.3892/ijmm.2017.3241
  • Pages: 373-380
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor necrosis factor‑α‑mediated (TNF‑α) epithelial‑mesenchymal transition (EMT) is associated with distant metastasis in patients with colorectal cancer with poor prognosis. Although traditional herbal medicines have long been used to treat colorectal cancer, the incidence and mortality in patients with colorectal cancer has continued to increase. Danggui‑Sayuk‑Ga‑Osuyu‑Saenggang‑Tang (DSGOST) has long been used for treatment of chills, while few studies have reported its anticancer effect. This study aimed to demonstrate the inhibitory effect of DSGOST on TNF‑α‑mediated invasion and migration of colorectal cancer HCT116 cell lines. MTT was used to measure cell viability. Wound healing and Τranswell invasion assay were used to detect migration and invasion of cells, respectively. The intracellular localization of proteins of interest was assessed by immunocytochemistry. Western blotting was performed to determine the expression level of various proteins. A non‑toxic dose of DSGOST (50 µg/ml) on HCT116 cells was determined by MTT assay. Furthermore, DSGOST prevented the TNF‑α‑induced invasive phenotype in HCT116 cells. DSGOST inhibition of the invasive phenotype was also associated with increased expression of EMT markers. Furthermore, DSGOST treatment blocked TNF‑α‑induced migration and invasion of HCT116 cells. In addition, DSGOST treatment inhibited TNF‑α‑mediated nuclear translocation of Snail. DSGOST treatment also downregulated TNF‑α‑induced phosphorylation of AKT and glycogen synthase kinase‑3β. Therefore, the findings of the current study suggest that DSGOST exhibits anti‑migration and anti‑invasion effects in TNF‑α‑treated HCT116 human colorectal cells.

Introduction

Colorectal cancer is one of leading causes of cancer-associated mortality worldwide (1,2). Despite advances in detection and therapy in previous years, the incidence and mortality in patients with colorectal cancer is still increasing throughout the world (36). The major cause of mortality patients with colorectal cancer is distant metastasis to other organs, such as the liver (7), lung (8), brain (9) and bone (10). Chronic inflammation has emerged as a key contributor for development and metastasis of cancer, including colorectal carcinoma (11,12). Inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukins (ILs), including IL-8, IL-6 and IL-1β, promote cancer progression (13). Among them, TNF-α contributes to proliferation and metastasis of colorectal cancer cells and its increased expression is often correlated with poor prognosis of patients (14). Although TNF-α expression has been considered to be a double-edged sword in cancer, it has been suggested that targeting TNF-α may be a successful strategy for cancer treatment (14,15). Activation of the TNF-α pathway promotes nuclear translocation of Snail transcription factor proteins (16). This translocation promotes epithelial-mesenchymal transition (EMT), a process that is considered to be a prerequisite for metastasis in various types of cancer, including colorectal cancer (1721). Furthermore, Snail overexpression in patients with colorectal cancer has also been correlated with aggressiveness and poor prognosis (22,23). Therefore, inhibition of TNF-α-induced EMT is likely to be effective for treatment of colorectal cancer.

Protein kinase B (AKT), one of the mitogen-activated protein kinases (MAPKs), regulates proliferation, differentiation, development, transformation and apoptosis (24). Disturbance of phosphoinositide 3-kinase/AKT signaling is associated with cancer progression and poor prognosis (25). Deregulated AKT phosphorylates glycogen synthase kinase-3β (GSK-3β), followed by inactivation of GSK-3β (26). Inactivated GSK-3β fails to tag Snail protein for ubiquitination and subsequent degradation (26). As Snail stabilization is tightly regulated by AKT/GSK-3β pathways, targeting this pathway is a promising strategy for cancer treatment. It has been reported that various anticancer agents inhibit tumor growth via suppressing AKT/GSK-3β/Snail signaling (2731).

Traditional herbal medicines have previously been researched for colorectal cancer treatment (3237). Danggui-Sayuk-Ga-Osuyu-Saenggang-Tang (DSGOST; Danggui-Sini-Jia-Wuzhuyu-Shengjian-Tang in Chinese, Tokishigyakukagoshuyushokyoto in Japanese) has long been used to cure patients suffered from Raynaud's syndrome (38,39), dysmenorrhea (40), atopic dermatitis (41), but not cancer. Choi et al (42) initially suggested a new use for DSGOST in the treatment of cancer as DSGOST was shown to inhibit pancreatic tumor growth and metastasis by suppressing vascular endothelial growth factor (VEGF)/VEGF receptor 2-dependent tumor angiogenesis. Furthermore, Nagata et al (43) demonstrated the inhibitory effect of DSGOST on growth of gastric cancer cells. Taken together, we hypothesize that DSGOST may be beneficial for cancer treatment.

In present study, the anticancer effect of DSOGST on TNF-α-mediated metastatic phenotypes was examined in HCT116 colorectal cancer cells. A non-cytotoxic dose of DSGOST inhibited TNF-α-induced migration and invasion of HCT116 cells. Furthermore, TNF-α induced morphological changes with either downregulation of E-cadherin or upregulation of N-cadherin whereas DSGOST reversed TNF-α-mediated changes. Furthermore, DSGOST suppressed TNF-α-induced nuclear translocation of Snail via inhibition of AKT/GSK-3β pathways. Therefore, these results demonstrated for the first time, the inhibitory effect of DSGOST on TNF-α-induced migration and invasion in HCT116 cells.

Materials and methods

Preparation of DSGOST

DSGOST powder was prepared as described previously (44). DSGOST consists of following herbal drugs; 1 g Angelica radix, 1 g Cinnamomi cortex, 1 g Paeoniae root, 1 g Akebia root, 0.67 g of Asarum, 0.67 g Glycyrrhiza, 1.67 g Zizyphus jujuba, 0.67 g Evodia fruit and 1.33 g ginger root. The dried mixture was dissolved in distilled water and stored at −80°C until use.

Cell culture and cell viability assay

HCT116 human colorectal cancer cell lines were purchased from Korean Cell Line Bank (Seoul, Korea) and cultured in RPMI-1640 medium (Welgene Inc., Daegu, Korea)supplemented with 10% fetal bovine serum (FBS; JR Scientific, Woodland, CA, USA) and 1% penicillin/streptomycin solution. For cell viability assay, HCT116 cells were seeded at the density of 4,000 cells/well in 96-wells plate and then treated with DSGOST (0, 10, 50 and 100 µg/ml). After 72 h incubation, cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay with an absorbance 570 nm.

Migration and invasion assay

For a cell migration assay, cells were cultured in 6-wells plates until 100% confluence and then the monolayer was scratched the monolayer with a 200 µl pipette tip. Cells were then pretreated with DSGOST for 2 h before incubation with TNF-α (R&D Systems, Minneapolis, MN, USA) for 48 h. The migrated area was analyzed by ImageJ software (64-bit Java 1.6.0_24; National Institutes of Health, Bethesda, MD, USA) and the average was calculated from independent experiments. For the invasion assay, Matrigel was mixed with coating buffer [0.01 M Tris (pH 8.0), 0.7% NaCl] at 1:1 ratio and then added in upper side of BD Transwell chamber in 24-well plates for overnight incubation. HCT116 (2×105 cells) were pre-incubated with 50 µg/ml DSGOST for 2 h, followed by 20 ng/ml of TNF-α for 48 h in the upper chamber. Culture medium with 10% FBS as chemoattractant was added into the lower chamber. After 48 h incubation, the cells were fixed with 4% paraformaldehyde for 5 min and then permeabilized with 100% methanol for 20 min. Non-invading cells were removed by cotton swab and the cells located at the underside of chamber were stained with 0.05% crystal violet for 20 min at room temperature. The four areas were randomly selected and cells were counted under a phase-contrast microscope.

Cellular fractionation

Cells were pretreated with DSGOST for 2 h, followed by incubation with TNF-α for 6 h. Cell lysates were suspended in 500 µl hypotonic buffer (20 mM Tris-HCl, pH 7.4, 10 mM NaCl, 3 mM MgCl2) and incubated on ice for 15 min, followed by centrifugation at 1,000 × g at 4°C. The supernatant (cytoplasmic fraction) was stored at -80°C until use. The obtained pellets were suspended in 50 µl cell extraction buffer containing 100 mM Tris (pH 7.4), 2 mM Na3VO4, 100 mM NaCl, 1% Triton X-100, 1 mM ethylenediaminetetraacetic acid (EDTA), 10% glycerol, 1 mM EGTA, 0.1% sodium dodecyl sulfate (SDS), 1 mM NaF, 0.5% deoxycholate and 20 mM Na4P2O7. After incubation on ice for 15 min, the mixture was centrifuged at 14,000 × g at 4°C. The supernatant (nuclear fraction) was stored at -80°C until use.

Western blotting

Cells were collected by scraping and then lysed with RIPA buffer containing 50 mM Tris-HCl, (pH 7.5), 150 mM NaCl, 1% Triton X-100, 2 mM EDTA, 0.1% SDS and 1% sodium deoxycholate. Equal amount of proteins [20 µg per well; protein concentration was determined using a Bio-Rad Bradford protein assay (Bio-Rad, Hercules, CA, USA)] were separated by 10–12% SDS-PAGE, followed by transfer to nitrocellulose membranes. After blocking with 2% skim milk in PBS-Tween at room temperature for 1 h, membranes were incubated with the appropriate antibodies at 4°C overnight. Anti-E-cadherin (cat. no. 3195, 1:1,000), tight junction protein-1 (ZO-1; cat. no. 8193; 1:500), N-cadherin (cat. no. 4061; 1:1,000), Snail (cat. no. 3879, 1:1,000), claudin-1 (cat. no. 13255; 1:500), p-AKT (cat. no. 9271; 1:500) and p-GSK-3β (cat. no. 9336; 1:500) antibodies were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Anti-lamin B (cat. no. sc6216; 1:500) and β-actin (cat. no. sc47778; 1:2,000) antibodies were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Horseradish peroxidase-conjugated secondary antibodies for mouse (cat. no. 7076; 1:1,000–3,000) and rabbit (cat. no. 7074; 1:1,000-2,000) were purchased from Cell Signaling Technology, Inc. The membranes were incubated with secondary antibodies for 1 h at room temperature. An enhanced chemiluminescence kit (DoGen, Seoul, Korea) was used for detection of HRP signal. ImageJ software (64-bit Java 1.6.0_24; National Institutes of Health) was used for densitometry.

Immunocytochemistry

The cells (80% confluency) were cultured on coverslip in 6-well plates with serum-free medium for 12 h and then pretreated with or without DSGOST. After 2 h incubation with DSGOST, cells were treated with TNF-α in medium with 1% FBS. Cells were washed with cold-PBS, fixed with 4% paraformaldehyde for 30 min, permeabilized with 0.1% Triton X-100 for 15 min and blocked with 2% bovine serum albumin (BSA) for 60 min at room temperature. Cells were washed with 0.5% BSA and were incubated with antibodies against E-cadherin (cat. no. 3195; 1:50), N-cadherin (cat. no. 4061; 1:50) and Snail (cat. no. 3879; 1:50) (Cell Signaling Technology, Inc.) at 4°C overnight. Following incubation, cells were incubated with Alexa Fluor 488-conjugated goat anti-rabbit IgG (cat. no. A-11008; 1:1,000; Thermo Fisher Scientific, Inc., Waltham, MA, USA) secondary antibodies for 1 h at room temperature. Nuclei were stained with TO-PRO-3 iodide (Thermo Fisher Scientific, Inc.). Confocal images were acquired with Zeiss LSM5 PASCAL confocal laser scanning microscope system (Carl Zeiss AG, Oberkochen, Germany). Fluorescence intensities were measured under the same condition for all experiments.

Statistical analysis

Data are presented as the mean ± standard error or standard deviation from at least three experiments with three replicates per each experiment. The statistical differences of means between the groups were analyzed by one-way analysis of variance with post-hoc Tukey test using SPSS software version 22.0 (IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

DSGOST inhibits TNF‑α‑induced EMT in HCT116 cells

To avoid interference resulting from the anti-proliferative effect of DSGOST on HCT116 cells on HCT116 cells, the effect of DSGOST on proliferation of cells was examined by an MTT assay. DSGOST (50 µg/ml) did not affect on the proliferation of cells (Fig. 1A). Thus, the effect of DSGOST (50 µg/ml) on TNF-α-mediated changes in morphology of cells was examined. While the cells in the TNF-α treatment group exhibited an invasive phenotype with a mesenchymal morphology, DSGOST treatment abrogated this change (Fig. 1B).

DSGOST reverses TNF‑α‑mediated EMT in HCT116 cells

TNF-α-induced invasive phenotype is associated with induction of EMT in colorectal cancer cells (16,45). Whether DSGOST prevents TNF-α-mediated changes in EMT markers was examined. Immunocytochemistry data demonstrated that TNF-α treatment induced mesenchymal morphology with decreased E-cadherin expression and increased N-cadherin expression, whereas pretreatment of DSGOST inhibited TNF-α-mediated responses (Fig. 2A). During EMT, epithelial cells acquire a mesenchymal-like phenotype, including the loss of E-cadherin-mediated cell-cell adhesion and the upregulation of N-cadherin. In TNF-α-treated HCT116 cells, epithelial cell markers E-cadherin and ZO-1 were decreased while mesenchymal markers Snail, N-cadherin and claudin-1 were increased. Additionally, DSGOST treatment reversed TNF-α-mediated protein expression (Fig. 2B and C). Thus, DSGOST is may inhibit TNF-α-mediated EMT in HCT116 cells.

DSGOST suppresses TNF‑α‑promoted migration and invasion of HCT116 cells

EMT process has been considered to be a prerequisite for migration and invasion of various cell types (46). As DSGOST inhibited TNF-α-induced EMT in HCT116 cells, it was also examined whether DSGOST suppresses TNF-α-mediated migration and invasion using wound healing assay and Transwell invasion assays, respectively. TNF-α increased migration and invasion of HCT116 cells, and DSGOST treatment blocked TNF-α-mediated migration and invasion (Figs. 3 and 4). Therefore, DSGOST is may suppress TNF-α-induced migration and invasion of HCT116 cells.

DSGOST inhibits TNF‑α‑induced nuclear translocation of Snail via inhibition of AKT/GSK‑3β pathways in HCT116 cells

TNF-α activates either the expression or nuclear trans-location of Snail protein, resulting in induction of EMT (16). As presented in Figs. 5 and 6, TNF-α promoted nuclear trans-location of Snail protein, whereas pretreatment with DSGOST prevented this translocation. Thus, DSGOST inhibits TNF-α-mediated EMT by suppressing Snail translocation into nucleus. As Snail is tightly regulated by GSK-3β, which is phosphorylated and inactivated by AKT (16,47), it was also further examined whether DSGOST inhibits TNF-α-mediated AKT/GSK-3β signaling. While TNF-α induced the phosphorylation of both AKT and GSK-3β, DSGOST inhibited their phosphorylation (Fig. 6B). Therefore, data indicated that DSGOST inhibition of TNF-α-induced Snail nuclear translocation is mediated by blocking the AKT/GSK-3β pathways.

Discussion

While DSGOST has been researched for the treatment of various disorders, recent studies have suggested DSGOST as a novel candidate for treatment of cancer (42,43). Thus, we hypothesized that DSGOST would be an effective treatment for colorectal cancer. Since TNF-α activates the EMT process in colorectal cancer cells, resulting in tumor metastasis, the inhibitory effect of DSGOST treatment on TNF-α-mediated migration and invasion of HCT116 cells was investigated in the current study. The findings showed that DSGOST suppressed TNF-α-mediated migration and invasion. Furthermore, DSGOST also blocked TNF-α-mediated EMT via inhibiting AKT/GSK-3β/Snail signaling.

In the late 1970s, TNF-α was considered to be an anticancer agent by suppressing tumor cell proliferation (48,49). On the other hand, several in vitro studies have reported the tumor promoting effect of TNF-α (16,50,51). Furthermore, clinical studies have reported that TNF-α expression levels in serum and tissues were elevated in patients with tumors (5255), suggesting that TNF-α may be a prognostic marker in patients with cancer. While the role of TNF-α in tumor malignancy is still controversial, the effect of recombinant TNF-α on EMT in HCT116 cells was investigated in the current study. The data showed that TNF-α treatment induced EMT phenotype. Furthermore, TNF-α also promoted migration and invasion. Although the cell type, TNF-α concentration and incubation period of TNF-α in the present study were limited, and the precise mechanisms of the effects remain to be demonstrated, the data provide evidence that TNF-α may contribute to tumor metastasis by inducing EMT.

The role of Snail in EMT has been validated in several cancer types (1820,22). Poor clinical outcomes of patients with colorectal cancer are associated with Snail overexpression (22,23). Wang et al (16) reported that TNF-α-induced EMT is regulated by Snail protein in at least two types of colorectal cancer cells, HCT116 and Caco-2. Furthermore, the same study clearly showed that AKT/GSK-3β signaling is responsible for TNF-α-mediated Snail stabilization in those cells, while inhibitors of nuclear factor-κB (NF-κB), MAPK and p38 failed to abrogate TNF-α-induced Snail expression (16). Since Snail stabilization is tightly regulated by AKT/GSK-3β signaling pathways, DSGOST inhibition of AKT/GSK-3β signaling indicates that one of the potential biological mechanisms of DSGOST inhibition of TNF-α-mediated EMT in HCT116 colorectal cancer cells is via blocking of AKT/GSK-3β/Snail signaling.

The potential effectiveness of multi-target approaches in cancer therapy has been emphasized previously (56). Phytochemicals and herbal mixtures, which may act on multiple targets, have anticancer activity (5766). Genistein, a dietary component with an anticancer effects against breast and prostate cancer, has been shown to regulate apoptosis, cell cycle, and NF-κB and AKT pathways (58). Proanthocyanidins have been reported to exhibit anticancer effects via antioxidative and anti-inflammatory properties (59). Ginsenoside, from ginseng plants, modulates the immune system, apoptosis and metastasis (57). Traditional Chinese medicine theory-based multi-herb prescriptions have benefits in the treatment of diseases including breast (60) and lung (61) cancer. SH003, a herbal prescription composed of Astragalus membranaceus, Angelica gigas and Trichosanthes kirilowii Maximowicz, exerts inhibitory effects against tumor growth by targeting apoptosis, autophagy and angiogenesis in several cancer types (62,6466). A previous study reported the antitumor and anti-angiogenesis effects of DSGOST (42). The present study also demonstrated that DSGOST inhibits TNF-α-mediated invasion and migration of HCT116 cells. Since both invasive phenotype and tumor angiogenesis contribute to tumor metastasis and poor prognosis of patients with cancer, we hypothesize that the multi-target properties of DSGOST may be useful for improving the prognosis of patients with cancer by dual inhibition of invasion and tumor angiogenesis. However, further in vitro and in vivo studies are required to demonstrate the anticancer property of DSGOST with a clear biological mechanism in several cancer types.

In conclusion, the data of the current study demonstrates, that DSGOST inhibits TNF-α-mediated EMT via suppressing AKT/GSK-3β/Snail pathways. Although the precise mechanism of DSGOST remains unclear, the results of the present study suggests that DSGOST is potentially beneficial for treating colorectal cancer. Furthermore, taken together with previous results indicating DSGOST inhibition of tumor angiogenesis, this suggests that DSGOST may be a novel multi-target herbal medicine for inhibition of tumor metastasis.

Acknowledgments

This study was supported by a grant from Korean Medicine R&D Project of the Ministry of Health and Welfare (grant no. B110043).

References

1 

Siegel R, Desantis C and Jemal A: Colorectal cancer statistics, 2014. CA Cancer J Clin. 64:104–117. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Pourhoseingholi MA: Increased burden of colorectal cancer in Asia. World J Gastrointest Oncol. 4:68–70. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Tournigand C, André T, Achille E, Lledo G, Flesh M, Mery-Mignard D, Quinaux E, Couteau C, Buyse M, Ganem G, et al: FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: A randomized GERCOR study. J Clin Oncol. 22:229–237. 2004. View Article : Google Scholar

5 

Van Cutsem E, Köhne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, D'Haens G, Pintér T, Lim R, Bodoky G, et al: Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 360:1408–1417. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Adam R, Avisar E, Ariche A, Giachetti S, Azoulay D, Castaing D, Kunstlinger F, Levi F and Bismuth F: Five-year survival following hepatic resection after neoadjuvant therapy for nonresectable colorectal. Ann Surg Oncol. 8:347–353. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Hamilton TD, Leugner D, Kopciuk K, Dixon E, Sutherland FR and Bathe OF: Identification of prognostic inflammatory factors in colorectal liver metastases. BMC Cancer. 14:5422014. View Article : Google Scholar : PubMed/NCBI

8 

Weiss L, Grundmann E, Torhorst J, Hartveit F, Moberg I, Eder M, Fenoglio-Preiser CM, Napier J, Horne CH, Lopez MJ, et al: Haematogenous metastatic patterns in colonic carcinoma: An analysis of 1541 necropsies. J Pathol. 150:195–203. 1986. View Article : Google Scholar : PubMed/NCBI

9 

Noura S, Ohue M, Shingai T, Fujiwara A, Imada S, Sueda T, Yamada T, Fujiwara Y, Ohigashi H, Yano M, et al: Brain metastasis from colorectal cancer: Prognostic factors and survival. J Surg Oncol. 106:144–148. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Jimi S, Yasui T, Hotokezaka M, Shimada K, Shinagawa Y, Shiozaki H, Tsutsumi N and Takeda S: Clinical features and prognostic factors of bone metastases from colorectal cancer. Surg Today. 43:751–756. 2013. View Article : Google Scholar

11 

Terzic J, Grivennikov S, Karin E and Karin M: Inflammation and colon cancer. Gastroenterology. 138:2101–2114 e2105. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Balkwill FR and Mantovani A: Cancer-related inflammation: Common themes and therapeutic opportunities. Semin Cancer Biol. 22:33–40. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Klampfer L: Cytokines, inflammation and colon cancer. Curr Cancer Drug Targets. 11:451–464. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Balkwill F: Tumour necrosis factor and cancer. Nat Rev Cancer. 9:361–371. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Balkwill F: TNF-alpha in promotion and progression of cancer. Cancer Metastasis Rev. 25:409–416. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Wang H, Wang HS, Zhou BH, Li CL, Zhang F, Wang XF, Zhang G, Bu XZ, Cai SH and Du J: Epithelial-mesenchymal transition (EMT) induced by TNF-α requires AKT/GSK-3β-mediated stabilization of snail in colorectal cancer. PLoS One. 8:e566642013. View Article : Google Scholar

17 

Peinado H, Olmeda D and Cano A: Snail, Zeb and bHLH factors in tumour progression: An alliance against the epithelial phenotype? Nat Rev Cancer. 7:415–428. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Dong C, Wu Y, Yao J, Wang Y, Yu Y, Rychahou PG, Evers BM and Zhou BP: G9a interacts with Snail and is critical for Snail-mediated E-cadherin repression in human breast cancer. J Clin Invest. 122:1469–1486. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Pon YL, Zhou HY, Cheung AN, Ngan HY and Wong AS: p70 S6 kinase promotes epithelial to mesenchymal transition through snail induction in ovarian cancer cells. Cancer Res. 68:6524–6532. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Wang J, Zhu X, Hu J, He G, Li X, Wu P, Ren X, Wang F, Liao W, Liang L, et al: The positive feedback between Snail and DAB2IP regulates EMT, invasion and metastasis in colorectal cancer. Oncotarget. 6:27427–27439. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Bates RC and Mercurio AM: The epithelial-mesenchymal transition (EMT) and colorectal cancer progression. Cancer Biol Ther. 4:365–370. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Kroepil F, Fluegen G, Vallböhmer D, Baldus SE, Dizdar L, Raffel AM, Hafner D, Stoecklein NH and Knoefel WT: Snail1 expression in colorectal cancer and its correlation with clinical and pathological parameters. BMC Cancer. 13:1452013. View Article : Google Scholar : PubMed/NCBI

23 

Kwon CH, Park HJ, Choi JH, Lee JR, Kim HK, Jo HJ, Kim HS, Oh N, Song GA and Park DY: Snail and serpinA1 promote tumor progression and predict prognosis in colorectal cancer. Oncotarget. 6:20312–20326. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Zhang W and Liu HT: MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 12:9–18. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Fresno Vara JA, Casado E, de Castro J, Cejas P, Belda-Iniesta C and González-Barón M: PI3K/Akt signalling pathway and cancer. Cancer Treat Rev. 30:193–204. 2004. View Article : Google Scholar : PubMed/NCBI

26 

McCubrey JA, Steelman LS, Bertrand FE, Davis NM, Sokolosky M, Abrams SL, Montalto G, D'Assoro AB, Libra M, Nicoletti F, et al: GSK-3 as potential target for therapeutic intervention in cancer. Oncotarget. 5:2881–2911. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Pappalardo F, Russo G, Candido S, Pennisi M, Cavalieri S, Motta S, McCubrey JA, Nicoletti F and Libra M: Computational modeling of PI3K/AKT and MAPK signaling pathways in melanoma cancer. PLoS One. 11:e01521042016. View Article : Google Scholar : PubMed/NCBI

28 

Ciuffreda L, McCubrey JA and Milella M: Signaling intermediates (PI3K/PTEN/AKT/mTOR and RAF/MEK/ERK pathways) as therapeutic targets for anticancer and anti-angiogenesis treatments. Curr Signal Transduct Ther. 4:130–143. 2009. View Article : Google Scholar

29 

El Touny LH and Banerjee PP: Akt GSK-3 pathway as a target in genistein-induced inhibition of TRAMP prostate cancer progression toward a poorly differentiated phenotype. Carcinogenesis. 28:1710–1717. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Bocca C, Bozzo F, Bassignana A and Miglietta A: Antiproliferative effects of COX-2 inhibitor celecoxib on human breast cancer cell lines. Mol Cell Biochem. 350:59–70. 2011. View Article : Google Scholar

31 

Firdous AB, Sharmila G, Balakrishnan S, RajaSingh P, Suganya S, Srinivasan N and Arunakaran J: Quercetin, a natural dietary flavonoid, acts as a chemopreventive agent against prostate cancer in an in vivo model by inhibiting the EGFR signaling pathway. Food Funct. 5:2632–2645. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Yim NH, Jung YP, Kim A, Ma CJ, Cho WK and Ma JY: Oyaksungisan, a traditional herbal formula, inhibits cell proliferation by induction of autophagy via JNK activation in human colon cancer cells. Evid Based Complement Alternat Med. 2013:2318742013. View Article : Google Scholar : PubMed/NCBI

33 

Hou F, Li W, Shi Q, Li H, Liu S, Zong S, Ren J, Chai J and Xu J: Yi Ai Fang, a traditional Chinese herbal formula, impacts the vasculogenic mimicry formation of human colorectal cancer through HIF-1α and epithelial mesenchymal transition. BMC Complement Altern Med. 16:4282016. View Article : Google Scholar

34 

Ohnishi Y, Fujii H, Hayakawa Y, Sakukawa R, Yamaura T, Sakamoto T, Tsukada K, Fujimaki M, Nunome S, Komatsu Y, et al: Oral administration of a Kampo (Japanese herbal) medicine Juzen-taiho-to inhibits liver metastasis of colon 26-L5 carcinoma cells. Jpn J Cancer Res. 89:206–213. 1998. View Article : Google Scholar : PubMed/NCBI

35 

Leung WK, Wu JC, Liang SM, Chan LS, Chan FK, Xie H, Fung SS, Hui AJ, Wong VW, Che CT, et al: Treatment of diarrhea-predominant irritable bowel syndrome with traditional Chinese herbal medicine: A randomized placebo-controlled trial. Am J Gastroenterol. 101:1574–1580. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Hosokawa A, Ogawa K, Ando T, Suzuki N, Ueda A, Kajiura S, Kobayashi Y, Tsukioka Y, Horikawa N, Yabushita K, et al: Preventive effect of traditional Japanese medicine on neurotoxicity of FOLFOX for metastatic colorectal cancer: A multicenter retrospective study. Anticancer Res. 32:2545–2550. 2012.PubMed/NCBI

37 

Yoshikawa K, Shimada M, Nishioka M, Kurita N, Iwata T, Morimoto S, Miyatani T, Komatsu M, Kashihara H and Mikami C: The effects of the Kampo medicine (Japanese herbal medicine) 'Daikenchuto' on the surgical inflammatory response following laparoscopic colorectal resection. Surg Today. 42:646–651. 2012. View Article : Google Scholar

38 

Tsukada R, Yamaguchi T, Hang L, Iseki M, Kobayashi H and Inada E: Effect of a traditional Japanese medicine goshajinkigan, tokishigyakukagoshuyushokyoto on the warm and cold sense threshold and peripheral blood flow. Health (NY). 6:757–763. 2014. View Article : Google Scholar

39 

Yoko K, Akira T and Hiroshi S: Efficacy of Kampo formula Tokishigyakukagoshuyushokyoto for cold syndrome evaluated with a novel clinical method using a patient-based questionnaire database. Kampo Med. 63:299–304. 2012. View Article : Google Scholar

40 

Oya A, Oikawa T, Nakai A, Takeshita T and Hanawa T: Clinical efficacy of Kampo medicine (Japanese traditional herbal medicine) in the treatment of primary dysmenorrhea. J Obstet Gynaecol Res. 34:898–908. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Hiromi K, Hisashi T, Shigeto Y, Takeshi N, Nobuyuki M, Daisuke T and Masamitsu I: Usefulness of Kampo formulas in the treatment of atopic dermatitis. J Tradit Medicines. 29:93–96. 2012.

42 

Choi HS, Lee K, Kim MK, Lee KM, Shin YC, Cho SG and Ko SG: DSGOST inhibits tumor growth by blocking VEGF/VEGFR2-activated angiogenesis. Oncotarget. 7:21775–21785. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Nagata T, Toume K, Long LX, Hirano K, Watanabe T, Sekine S, Okumura T, Komatsu K and Tsukada K: Anticancer effect of a Kampo preparation Daikenchuto. J Nat Med. 70:627–633. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Lee K, Cho SG, Woo SM, Kim AJ, Lee KM, Go HY, Sun SH, Kim TH, Jung KY, Choi YK, et al: Danggui Sayuk Ga Osuyu Senggang Tang ameliorates cold induced vasoconstriction in vitro and in vivo. Mol Med Rep. 14:4723–4728. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Bates RC and Mercurio AM: Tumor necrosis factor-alpha stimulates the epithelial-to-mesenchymal transition of human colonic organoids. Mol Biol Cell. 14:1790–1800. 2003. View Article : Google Scholar : PubMed/NCBI

46 

Christiansen JJ and Rajasekaran AK: Reassessing epithelial to mesenchymal transition as a prerequisite for carcinoma invasion and metastasis. Cancer Res. 66:8319–8326. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Zhou BP, Deng J, Xia W, Xu J, Li YM, Gunduz M and Hung MC: Dual regulation of Snail by GSK-3 beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat Cell Biol. 6:931–940. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Green S, Dobrjansky A and Chiasson MA: Murine tumor necrosis-inducing factor: Purification and effects on myelomonocytic leukemia cells. J Natl Cancer Inst. 68:997–1003. 1982.PubMed/NCBI

49 

Carswell EA, Old LJ, Kassel RL, Green S, Fiore N and Williamson B: An endotoxin-induced serum factor that causes necrosis of tumors. Proc Natl Acad Sci USA. 72:3666–3670. 1975. View Article : Google Scholar : PubMed/NCBI

50 

Wu Y and Zhou BP: TNF-alpha/NF-kappaB/Snail pathway in cancer cell migration and invasion. Br J Cancer. 102:639–644. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Zins K, Abraham D, Sioud M and Aharinejad S: Colon cancer cell-derived tumor necrosis factor-alpha mediates the tumor growth-promoting response in macrophages by upregulating the colony-stimulating factor-1 pathway. Cancer Res. 67:1038–1045. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Ferrajoli A, Keating MJ, Manshouri T, Giles FJ, Dey A, Estrov Z, Koller CA, Kurzrock R, Thomas DA, Faderl S, et al: The clinical significance of tumor necrosis factor-alpha plasma level in patients having chronic lymphocytic leukemia. Blood. 100:1215–1219. 2002.PubMed/NCBI

53 

Csiszár A, Szentes T, Haraszti B, Balázs A, Petrányi GG and Pócsik E: The pattern of cytokine gene expression in human colorectal carcinoma. Pathol Oncol Res. 10:109–116. 2004. View Article : Google Scholar : PubMed/NCBI

54 

Grimm M, Lazariotou M, Kircher S, Höfelmayr A, Germer CT, von Rahden BH, Waaga-Gasser AM and Gasser M: Tumor necrosis factor-α is associated with positive lymph node status in patients with recurrence of colorectal cancer-indications for anti-TNF-α agents in cancer treatment. Cell Oncol (Dordr). 34:315–326. 2011. View Article : Google Scholar

55 

Al Obeed OA, Alkhayal KA, Al Sheikh A, Zubaidi AM, Vaali-Mohammed MA, Boushey R, Mckerrow JH and Abdulla MH: Increased expression of tumor necrosis factor-α is associated with advanced colorectal cancer stages. World J Gastroenterol. 20:18390–18396. 2014. View Article : Google Scholar

56 

Petrelli A and Giordano S: From single- to multi-target drugs in cancer therapy: When aspecificity becomes an advantage. Curr Med Chem. 15:422–432. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Attele AS, Wu JA and Yuan CS: Ginseng pharmacology: Multiple constituents and multiple actions. Biochem Pharmacol. 58:1685–1693. 1999. View Article : Google Scholar : PubMed/NCBI

58 

Banerjee S, Li Y, Wang Z and Sarkar FH: Multi-targeted therapy of cancer by genistein. Cancer Lett. 269:226–242. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Nandakumar V, Singh T and Katiyar SK: Multi-targeted prevention and therapy of cancer by proanthocyanidins. Cancer Lett. 269:378–387. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Gao JL, He TC, Li YB and Wang YT: A traditional Chinese medicine formulation consisting of Rhizoma Corydalis and Rhizoma Curcumae exerts synergistic antitumor activity. Oncol Rep. 22:1077–1083. 2009.PubMed/NCBI

61 

Di Maio M, Costanzo R, Giordano P, Piccirillo MC, Sandomenico C, Montanino A, Carillio G, Muto P, Jones DR, Daniele G, et al: Integrated therapeutic approaches in the treatment of locally advanced non-small cell lung cancer. Anticancer Agents Med Chem. 13:844–851. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Choi YJ, Choi YK, Lee KM, Cho SG, Kang SY and Ko SG: SH003 induces apoptosis of DU145 prostate cancer cells by inhibiting ERK-involved pathway. BMC Complement Altern Med. 16:5072016. View Article : Google Scholar : PubMed/NCBI

63 

Choi EK, Kim SM, Hong SW, Moon JH, Shin JS, Kim JH, Hwang IY, Jung SA, Lee DH, Lee EY, et al: SH003 selectively induces p73 dependent apoptosis in triple negative breast cancer cells. Mol Med Rep. 14:3955–3960. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Choi YK, Cho SG, Choi YJ, Yun YJ, Lee KM, Lee K, Yoo HH, Shin YC and Ko SG: SH003 suppresses breast cancer growth by accumulating p62 in autolysosomes. Oncotarget. Aug 19–2016.Epub ahead of print.

65 

Choi HS, Kim MK, Lee K, Lee KM, Choi YK, Shin YC, Cho SG and Ko SG: SH003 represses tumor angiogenesis by blocking VEGF binding to VEGFR2. Oncotarget. 7:32969–32979. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Choi YK, Cho SG, Woo SM, Yun YJ, Park S, Shin YC and Ko SG: Herbal extract SH003 suppresses tumor growth and metastasis of MDA-MB-231 breast cancer cells by inhibiting STAT3-IL-6 signaling. Mediators Inflamm. 2014:4921732014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 41 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee K, Cho S, Choi YK, Choi Y, Lee G, Jeon C and Ko S: Herbal prescription, Danggui-Sayuk-Ga-Osuyu-Senggang-Tang, inhibits TNF-α-induced epithelial-mesenchymal transition in HCT116 colorectal cancer cells. Int J Mol Med 41: 373-380, 2018
APA
Lee, K., Cho, S., Choi, Y.K., Choi, Y., Lee, G., Jeon, C., & Ko, S. (2018). Herbal prescription, Danggui-Sayuk-Ga-Osuyu-Senggang-Tang, inhibits TNF-α-induced epithelial-mesenchymal transition in HCT116 colorectal cancer cells. International Journal of Molecular Medicine, 41, 373-380. https://doi.org/10.3892/ijmm.2017.3241
MLA
Lee, K., Cho, S., Choi, Y. K., Choi, Y., Lee, G., Jeon, C., Ko, S."Herbal prescription, Danggui-Sayuk-Ga-Osuyu-Senggang-Tang, inhibits TNF-α-induced epithelial-mesenchymal transition in HCT116 colorectal cancer cells". International Journal of Molecular Medicine 41.1 (2018): 373-380.
Chicago
Lee, K., Cho, S., Choi, Y. K., Choi, Y., Lee, G., Jeon, C., Ko, S."Herbal prescription, Danggui-Sayuk-Ga-Osuyu-Senggang-Tang, inhibits TNF-α-induced epithelial-mesenchymal transition in HCT116 colorectal cancer cells". International Journal of Molecular Medicine 41, no. 1 (2018): 373-380. https://doi.org/10.3892/ijmm.2017.3241