Pien Tze Huang inhibits metastasis of human colorectal carcinoma cells via modulation of TGF-β1/ZEB/miR‑200 signaling network

  • Authors:
    • Aling Shen
    • Wei Lin
    • Youqin Chen
    • Liya Liu
    • Hongwei Chen
    • Qunchuan Zhuang
    • Jiumao Lin
    • Thomas J. Sferra
    • Jun Peng
  • View Affiliations

  • Published online on: November 24, 2014     https://doi.org/10.3892/ijo.2014.2772
  • Pages: 685-690
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor metastasis, a complex process involving the spread of malignant tumor cells from a primary tumor site to a distant organ, is a major cause of failure of cancer chemotherapy. Epithelial-mesenchymal transition (EMT) is a critical step for the initiation of cancer metastasis. The processes of EMT and metastasis are highly regulated by a double-negative feedback loop consisting of TGF-β1/ZEB pathway and miR‑200 family, which therefore has become a promising target for cancer chemotherapy. Pien Tze Huang (PZH), a well‑known traditional Chinese formula first prescribed in the Ming Dynasty, has been demonstrated to be clinically effective in the treatment of various types of human malignancy including colorectal cancer (CRC). Our published data proposed that PZH was able to induce apoptosis, inhibit cell proliferation and tumor angiogenesis, leading to the suppression of CRC growth in vitro and in vivo. To further elucidate the mode of action of PZH, in the present study we evaluated its effects on the metastatic capacities of human colorectal carcinoma HCT-8 cells and investigated the underlying molecular mechanisms. We found that PZH significantly inhibited the migration and invasion of HCT-8 cells in a dose-dependent manner. In addition, PZH treatment inhibited the expression of key mediators of TGF-β1 signaling, such as TGF-β1, Smad2/3 and Smad4. Moreover, PZH treatment suppressed the expression of ZEB1 and ZEB2, two critical target genes of TGF-β1 pathway, leading to a decrease in the expression of mesenchymal marker N-cadherin and an increased expression of epithelial marker E-cadherin. Furthermore, PZH treatment upregulated the expression of miR‑200a, miR‑200b and miR‑200c. Collectively, our findings in this study suggest that PZH can inhibit metastasis of colorectal cancer cells via modulating TGF-β1/ZEB/miR‑200 signaling network, which might be one of the mechanisms whereby PZH exerts its anticancer function.

Introduction

Colorectal cancer (CRC) is one of the most common cancers and a leading cause of cancer-related deaths (1,2). To date, the mainstay of anti-CRC treatment includes surgery, chemotherapy and radiotherapy. However, due to tumor recurrence and metastasis the long-term survival and prognosis of patients remains quite poor (3,4). Tumor metastasis is a complex process involving the spread of malignant tumor cells from a primary tumor site to a distant organ, which is a major cause of failure of cancer treatment (57). Epithelial-mesenchymal transition (EMT) is a critical step for the initiation of cancer metastasis (8,9). The processes of EMT and metastasis are highly regulated by multiple mechanisms, including TGF-β1/ZEB pathways and miRNA 200 family (1014).

TGF-β1 is the prototypic member of transforming growth factor β superfamily. The activation of TGF-β signaling pathway is initiated by the binding of ligands to a type II receptor, resulting in the phosphorylation/activation of a type I receptor. The activated type I receptor then phosphorylates SMAD2/3 that in turn bind to SMAD4. The SMAD complex translocates to the nucleus to regulate the expression of target genes, including the ZEB (zinc finger E-box-binding homeobox) transcription factor family (15,16). Upon activation, ZEB transcription factors suppress epithelial marker gene expression and upregulate mesenchymal gene expression, leading to the processes of EMT and cancer metastasis (17,18). MicroRNAs (miRNA) are a class of endogenous short non-coding RNAs (19–24 nucleotides), which function primarily to negatively regulate target gene expression by specifically binding to the 3′-untranslational region (3′-UTR) of target mRNAs (1921). It has been shown that miRNAs function more likely as oncogenes or tumor suppressors to modulate multiple oncogenic cellular processes, such as cell proliferation, apoptosis and metastasis (2224). The miR-200 family members, including miR-200a, miR-200b and miR-200c, have been proposed to act as tumor suppressors that inhibit EMT by downregulating the expression of ZEB1 and ZEB2 (11,14,2527). However, the expression of miR-200 family is negatively regulated by TGF-β signaling, probably via TGF-β-induced DNA methylation of the miR-200 loci (12). Thus, TGF-β/ZEB/miR-200 signaling network creates a double-negative feedback loop that plays an essential role in the initiation of EMT and cancer metastasis; which therefore becomes a promising target for cancer chemotherapy (13,14).

Recently, traditional Chinese medicines (TCM) have received great interest in the field of anticancer treatment since they have fewer adverse effects as compared to modern chemotherapeutics and have been used in China for thousands of years as important alternative remedies for various diseases including cancer (28,29). Pien Tze Huang (PZH) is a well-known TCM formula that was first prescribed >450 years ago in the Ming Dynasty. The main ingredients of PZH include Moschus, Calculus Bovis, Snake Gall and Radix Notoginseng. These products together confer PZH properties of heat clearing, detoxification, dissipation of hard mass, detumescence and analgesia (30). Traditionally, PZH has been used to clinically treat traumatic injuries and a variety of inflammatory diseases, particularly hepatitis (3032). More importantly, PZH has also been used in China and Southeast Asia for centuries as a folk remedy for treatment of various types of human cancer. We recently reported that PZH can inhibit colon cancer growth through the promotion of cancer cell apoptosis, the inhibition of cell proliferation and tumor angiogenesis, which is probably mediated by modulation of multiple signaling pathways (3340). To further elucidate the mode of action of PZH, in the present study we evaluated its effects on the metastatic capacities of human colorectal carcinoma HCT-8 cells and investigated the underlying molecular mechanisms.

Materials and methods

Materials and reagents

Roswell Park Memorial Institute (RPMI)-1640 medium, fetal bovine serum (FBS), penicillin-streptomycin, were obtained from Life Technologies Corp. (Grand Island, NY, USA). N-cadherin and E-cadherin antibodies were purchased from Abcam (HK) Ltd. (Hong Kong, China). TGF-β1, SMAD2/3, SMAD4, ZEB1, ZEB2 and β-actin antibodies, horseradish peroxidase (HRP)-conjugated secondary antibodies were provided by Cell Signaling Technology (Beverly, MA, USA). Transwell chambers were obtained from Corning Life Sciences (Tewksbury, MA, USA). BD BioCoat Matrigel Invasion Chamber was purchased from BD Bioscience (San Jose, CA, USA). PrimeScript RT reagent kit, RNAiso for Small RNA kit and SYBR Premix Ex Taq II kit were provided by Dalian Takara Biotechnology Co., Ltd. (Dalian, Liaoning, China). All the other chemicals, unless otherwise stated, were obtained from Sigma Chemicals (St. Louis, MO, USA).

Preparations of PZH

PZH was obtained from, and authenticated by the sole manufacturer Zhangzhou Pien Tze Huang Pharmaceutical Co. Ltd., China (Chinese FDA approval no. Z35020242). Stock solutions of PZH were prepared just before use by dissolving the PZH powder in PBS (phosphate-buffered saline) to a concentration of 20 mg/ml. The working concentrations of PZH were made by diluting the stock solution in the culture medium.

Cell culture

Human colorectal carcinoma HCT-8 cells were obtained from Nanjing KeyGen Biotech. Co. Ltd. (Nanjing, Jiangsu, China). Cells were grown in RPMI-1640 medium containing 10% (v/v) FBS, 100 U/ml penicillin and 100 μg/ml streptomycin in a 37°C humidified incubator with 5% CO2. The cells were subcultured at 80–90% confluency.

Evaluation of cell migration by wound-healing assay

Migration of HCT-8 cells was examined by wound-healing assay. Cells were seeded into 6-well plate at a density of 1×106 cells/well in 2 ml medium. After 24 h of incubation, cells were scraped away vertically in each well by using a P100 pipette tip. Three randomly selected views along the scraped line were photographed on each well using a phase-contrast inverted microscope (Leica, Germany) at a magnification of ×100. Cells were then treated with indicated concentrations of PZH for 24 h, and another set of images were taken using the same method. A reduction in the scraped area indicates a sign of migration.

Measurement of cell migration and invasion by transwell assay

Migration assay was performed using transwell cell culture chambers with 8-μm pore filters (Corning Life Sciences, USA). After treatment with various concentrations of PZH for 24 h, HCT-8 cells were trypsinized and resuspended in serum-free RPMI-1640. A total of 5×104 cells in 200 μl of serum-free RPMI-1640 were plated in the upper chambers. RPMI-1640 media containing 10% (v/v) FBS was used in the lower chambers as a chemoattractant. Cells were allowed to migrate for 12 h, and the non-migrated cells were removed from the upper surface of transwell membranes by a cotton swab. Membranes were then stained with crystal violet. For quantification, the average number of migrating cells per field was assessed by counting 3 random fields under a phase-contrast microscope (Leica) at a magnification of ×200. For cell invasion assay, the procedure was the same as that of above-described migration analysis, except that the upper chambers were coated with Matrigel Matrix (BD Biosciences, USA).

Western blot analysis

HCT-8 cells were seeded into 25 cm2 flasks at a density of 1.5×106 cells/flask in 5 ml medium. After incubation for 24 h, the cells were treated with the indicated concentrations of PZH for 24 h. The treated cells were lysed with mammalian cell lysis buffer containing protease and phosphatase inhibitor cocktails. Total protein concentrations were determined by BCA assay. Equal amounts of total proteins were resolved in 12% SDS-PAGE gels and electroblotted. The PVDF membranes were blocked with 5% skimmed milk and probed with primary antibodies N-cadherin, E-cadherin, TGF-β1, SMAD2/3, SMAD4, ZEB1, ZEB2 and β-actin overnight at 4°C and subsequently with the appropriate HRP-conjugated secondary antibody followed by enhanced chemiluminescence detection.

Q-PCR analysis

Total small RNA from HCT-8 cells was isolated with RNAiso for Small RNA kit. Total small RNA (500 ng) was reverse-transcribed with SYBR PrimeScript miRNA RT-PCR kit according to the manufacturer’s instructions. The obtained cDNA was used to determine the miRNA amount of miR-200a, miR-200b and miR-200c, U6 was used as an internal control. The primers of miR-200a (DHM0178), miR-200b (DHM0179), miR-200c (DHM0180) and U6 (D356-03) were obtained from Dalian Takara Biotechnology Co., Ltd. Quantitative PCR was performed using SYBR Premix Ex Taq II in an ABI 7500 Fast instrument. Q-PCR reactions were carried out following the manufacturer’s protocol. miRNA expression values were determined as ΔCt=Ct (sample)−Ct (U6) and relative quantities between different samples were determined as ΔΔCt=ΔCt (sample 1)−ΔCt (sample 2), the values were expressed as 2−ΔΔCt. All Q-PCR reactions were conducted in triplicate.

Statistical analysis

The data are presented as the means of three determinations and was analyzed using the SPSS package for Windows (Version 18.0). Statistical analysis of the data was performed with Student’s t-test and ANOVA. Differences with P<0.05 were considered statistically significant.

Results

PZH inhibits migration and invasion of HCT-8 cells

We first performed a wound-healing assay to evaluate the effect of PZH on the migration of HCT-8 cells. As shown in Fig. 1, after post-wounding for 24 h, untreated HCT-8 cells migrated into the clear area, whereas PZH treatment dose-dependently inhibited HCT-8 cell migration. We further verified these results using transwell assay; and the data showed that treatment with 0.25–0.75 mg/ml of PZH for 24 h dose-dependently reduced cell migratory rate of HCT-8 cells by 44.4–85.8%, as compared to untreated cells (Fig. 2, P<0.05). We next determined the effect of PZH on the invasion capacity of HCT-8 cells using the transwell assay. As shown in Fig. 3, compared with untreated cells (100%), the invasion rate of HCT-8 cells following treatment with 0.25, 0.5 or 0.75 mg/ml of PZH was 46.0±8.4, 29.6±3.0 or 19.1±4.0%, respectively (P<0.05). Taken together, these data suggest that PZH can inhibit metastasis of human colorectal cancer cells.

PZH modulates the activation of TGF-β1 pathway and the expression of EMT-regulatory genes in HCT-8 cells

To determine the PZH effect on the activation of TGF-β1 signaling, we examined the protein expression of several key mediators of this pathway using western blot analysis. As shown in Fig. 4, the protein expression levels of TGF-β1, Smad2/3 and Smad4 were downregulated by PZH treatment in a dose-dependent manner. Moreover, PZH treatment suppressed the expression of TGF-β1 target genes ZEB1 and ZEB2, leading to the downregulation of expression of mesenchymal marker N-cadherin as well as an increase in the expression of epithelial marker E-cadherin (Fig. 4). Therefore, the inhibitory effect of PZH on cancer cell metastasis might be mediated by the suppression of TGF-β1 pathway and the process of EMT.

PZH upregulates the expression of miR-200a, miR-200b and miR-200c in HCT-8 cells

To further explore the mechanism of anti-metastasis activities of PZH, we determined the expression of miR-200 family in HCT-8 cells using Q-PCR assay. As shown in Fig. 5, PZH treatment significantly and dose-dependently increased the expression of miR-200a, miR-200b and miR-200c, consistent with the observations that PZH inhibited the TGF-β1 pathway and expression of ZEB transcription factors (Fig. 4).

Discussion

Drug resistance and intrinsic cytotoxicity against normal cells profoundly limit the long-term use of currently-used chemotherapeutic regimens and thereby their therapeutic effectiveness (41,42), emphasizing the need for the development of novel antitumor drugs. Due to the relatively higher safety and the long history of pharmacological applications, traditional Chinese medicines (TCM) have attracted great interest in the field of cancer treatment (28,29). TCM formula is a complex combination of many natural products, each of which contains numerous chemical compounds. Therefore, TCM formulas are considered to be multi-component and multi-target agents exerting their therapeutic function in a more holistic way; and discovering naturally-occurring agents could be a promising approach of cancer treatment. Pien Tze Huang (PZH) is a well-known TCM formula that has been used in China and Southeast Asia for centuries as a folk remedy for various types of cancer. We recently reported that PZH can inhibit colon cancer growth through the promotion of cancer cell apoptosis, the inhibition of cell proliferation and tumor angiogenesis, which is probably mediated by modulation of multiple signaling pathways (3340). These data demonstrate that PZH possesses a broad range of anticancer activities due to its ability to affect multiple intracellular targets, suggesting that PZH could be a novel multi-target anticancer agent.

Tumor metastasis is a complex process involving the spread of malignant tumor cells from a primary tumor site to a distant organ, which is a major cause of failure of clinical cancer chemotherapy and therefore has become an important focus for anticancer therapies (57,13,14). To further elucidate the mode of action of PZH, in the present study we evaluated its effects on cancer metastasis. Using wound healing and transwell assays we found that PZH treatment significantly inhibited the migration and invasion of human colorectal carcinoma HCT-8 cells in a dose-dependent manner, demonstrating the inhibitory activity of PZH on the metastatic capacities of colorectal cancer cells. Epithelial-mesenchymal transition (EMT) is a biological process in which epithelial cells lose their polarity and cell-cell adhesion, and acquire migratory and invasive properties of mesenchymal cells (8,9,14,15). After acquiring a mesenchymal phenotype through the process of EMT, carcinoma cells invade adjacent tissues, break through the basement membrane, and eventually enter the bloodstream leading to cancer metastasis (8,9,1417). Therefore, EMT is an essential step for the initiation of cancer metastasis. Using western blot analysis we found that PZH treatment reduced the protein expression of mesenchymal marker N-cadherin but increased that of epithelial marker E-cadherin, indicating that the anti-metastasis activity of PZH was associated with its inhibitory effect on EMT. The processes of EMT and metastasis are highly regulated by multiple mechanisms, including TGF-β1/SMAD pathways and miRNA 200 family (1014). The activation of TGF-β1 signaling enhances the expression of ZEB transcription factors, which in turn modulates the expression of EMT-regulatory genes resulting in the initiation of EMT (15,16). Interestingly, the expression of ZEB transcription factors can be downregulated by miR-200 family members (11,14,2527); but miR-200 family expression is negatively regulated by TGF-β1 signaling (12), forming a double-negative feedback loop to control the processes of EMT and metastasis (13,14). Data from western blot and Q-PCR analyses indicated that PZH suppressed the activation of TGF-β1 pathway and the expression of ZEB1 and ZEB2, whereas the expression of miR-200a, miR-200b and miR-200c was upregulated after PZH treatment.

In conclusion, here we report that PZH can inhibit the metastatic capacity of human colorectal carcinoma cells via modulating TGF-β1/ZEB/miR-200 signaling network, which might be one of the mechanisms whereby PZH exerts its anticancer function.

Acknowledgements

This study was supported by the National Natural Science Foundations of China (81373819 and 81202790) and the China Postdoctoral Science Foundation (2013T60636).

Abbreviations:

CRC

colorectal cancer

PZH

Pien Tze Huang

TCM

traditional Chinese medicine

TGF-β

transforming growth factor-β

EMT

epithelial-to-mesenchymal transition

ZEB

zinc finger E-box-binding homeobox

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 : PubMed/NCBI

2 

Markowitz SD and Bertagnolli MM: Molecular basis of colorectal cancer. N Engl J Med. 361:2449–2460. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Koyanagi K, Bilchik AJ, Saha S, Turner RR, Wiese D, McCarter M, Shen P, Deacon L, Elashoff D and Hoon DS: Prognostic relevance of occult nodal micrometastases and circulating tumor cells in colorectal cancer in a prospective multicenter trial. Clin Cancer Res. 14:7391–7396. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Bilchik AJ, Hoon DS, Saha S, Turner RR, Wiese D, DiNome M, Koyanagi K, McCarter M, Shen P, Iddings D, Chen SL, Gonzalez M, Elashoff D and Morton DL: Prognostic impact of micrometastases in colon cancer: interim results of a prospective multicenter trial. Ann Surg. 246:568–575. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Johnson SM, Gulhati P, Rampy BA, Han Y, Rychahou PG, Doan HQ, Weiss HL and Evers BM: Novel expression patterns of PI3K/Akt/mTOR signaling pathway components in colorectal cancer. J Am Coll Surg. 210:767–778. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Christofori G: New signals from the invasive front. Nature. 441:444–450. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Stein U and Schlag PM: Clinical, biological, and molecular aspects of metastasis in colorectal cancer. Recent Results Cancer Res. 176:61–80. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Pecina-Slaus N, Cicvara-Pecina T and Kafka A: Epithelial-to-mesenchymal transition: possible role in meningiomas. Front Biosci (Elite Ed). 13:889–896. 2012. View Article : Google Scholar

9 

Guarino M, Rubino B and Ballabio G: The role of epithelial-mesenchymal transition in cancer pathology. Pathology. 13:305–318. 2007. View Article : Google Scholar

10 

Xu Y and Pasche B: TGF-beta signaling alterations and susceptibility to colorectal cancer. Hum Mol Genet. 16:R14–R20. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Eades G, Yao Y, Yang M, Zhang Y, Chumsri S and Zhou Q: miR-200a regulates SIRT1 expression and epithelial to mesenchymal transition (EMT)-like transformation in mammary epithelial cells. J Biol Chem. 286:25992–26002. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Gregory PA, Bracken CP, Smith E, Bert AG, Wright JA, Roslan S, Morris M, Wyatt L, Farshid G, Lim YY, Lindeman GJ, Shannon MF, Drew PA, Khew-Goodall Y and Goodall GJ: An autocrine TGF-beta/ZEB/miR-200 signaling network regulates establishment and maintenance of epithelial-mesenchymal transition. Mol Biol Cell. 22:1686–1698. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Xiong M, Jiang L, Zhou Y, Qiu W, Fang L, Tan R, Wen P and Yang J: The miR-200 family regulates TGF-β1-induced renal tubular epithelial to mesenchymal transition through Smad pathway by targeting ZEB1 and ZEB2 expression. Am J Physiol Renal Physiol. 302:F369–F379. 2012. View Article : Google Scholar

14 

Hur K, Toiyama Y, Takahashi M, Balaguer F, Nagasaka T, Koike J, Hemmi H, Koi M, Boland CR and Goel A: MicroRNA-200c modulates epithelial-to-mesenchymal transition (EMT) in human colorectal cancer metastasis. Gut. 62:1315–1326. 2013. View Article : Google Scholar :

15 

Moustakas A and Heldin CH: Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 98:1512–1520. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Wu Y, Sato F, Yamada T, Bhawal UK, Kawamoto T, Fujimoto K, Noshiro M, Seino H, Morohashi S, Hakamada K, Abiko Y, Kato Y and Kijima H: The BHLH transcription factor DEC1 plays an important role in the epithelial-mesenchymal transition of pancreatic cancer. Int J Oncol. 41:1337–1346. 2012.PubMed/NCBI

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 

Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, Bruyneel E, Mareel M, Huylebroeck D and van Roy F: The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell. 7:1267–1278. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Valencia-Sachez MA, Liu J, Hannon GJ and Parker R: Control of translation and mRNA degradation by miRNAs and siRNAs. Genes Dev. 20:515–524. 2006. View Article : Google Scholar

20 

Bagga S and Pasquinelli AE: Identification and analysis of microRNAs. Genet Eng. 27:1–20. 2006. View Article : Google Scholar

21 

Bar N and Dikstein R: miR-22 forms a regulatory loop in PTEN/AKT pathway and modulates signaling kinetics. PLoS One. 5:e108592010. View Article : Google Scholar : PubMed/NCBI

22 

Leskelä S, Leandro-García LJ, Mendiola M, Barriuso J, Inglada-Pérez L, Muñoz I, Martínez-Delgado B, Redondo A, de Santiago J and Robledo M: The miR-200 family controls β-tubulin iii expression and is associated with paclitaxel-based treatment response and progression-free survival in ovarian cancer patients. Endocr Relat Cancer. 18:85–95. 2011. View Article : Google Scholar

23 

Uhlmann S, Zhang J, Schwäger A, Mannsperger H, Riazalhosseini Y, Burmester S, Ward A, Korf U, Wiemann S and Sahin Ö: MiR-200bc/429 cluster targets plcγ1 and differentially regulates proliferation and egf-driven invasion than miR-200a/141 in breast cancer. Oncogene. 29:4297–4306. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Li H, Tang J, Lei H, Cai P, Zhu H, Li B, Xu X, Xia Y and Tang W: Decreased MiR-200a/141 suppress cell migration and proliferation by targeting PTEN in Hirschsprung’s disease. Cell Physiol Biochem. 34:543–553. 2014. View Article : Google Scholar

25 

Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y and Goodall GJ: The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 10:593–601. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Park SM, Gaur AB, Lengyel E and Peter ME: The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 22:894–907. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Wang CH, Chen CL, More SV, Hsiao PW, Hung WC and Li WS: The tetraindole SK228 reverses the epithelial-to-mesenchymal transition of breast cancer cells by up-regulating members of the miR-200 family. PLoS One. 9:e1010882014. View Article : Google Scholar : PubMed/NCBI

28 

Gordaliza M: Natural products as leads to anticancer drugs. Clin Transl Oncol. 9:767–776. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Ji HF, Li XJ and Zhang HY: Natural products and drug discovery. EMBO Rep. 10:194–200. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Chinese Pharmacopoeia Commission. Pharmacopoeia of the Peoples Republic of China. 1. Chinese Medical Science and Technology Press; Beijing: pp. 573–575. 2010

31 

Lee KK, Kwong WH, Chau FT, Yew DT and Chan WY: Pien Tze Huang protects the liver against carbon tetrachloride-induced damage. Pharmacol Toxicol. 91:185–192. 2002. View Article : Google Scholar

32 

Chan WY, Chau FT, Lee KK, Kwong WH and Yew DT: Substitution for natural musk in Pien Tze Huang does not affect its hepatoprotective activities. Hum Exp Toxicol. 23:35–47. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Lin JM, Wei LH, Chen YQ, Liu XX, Hong ZF, Sferra TJ and Peng J: Pien Tze Huang-induced apoptosis in human colon cancer HT-29 cells is associated with regulation of the Bcl-2 family and activation of caspase 3. Chin J Integr Med. 17:685–690. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Zhuang QC, Hong F, Shen AL, Zheng LP, Zeng JW, Lin W, Chen YQ, Sferra TJ, Hong ZF and Peng J: Pien Tze Huang inhibits tumor cell proliferation and promotes apoptosis via suppressing the STAT3 pathway in colorectal cancer mouse. Int J Oncol. 26:1569–1574. 2012.

35 

Shen AL, Hong F, Liu LY, Lin JM, Zhuang QC, Hong ZF and Peng J: Effects of Pien Tze Huang on angiogenesis in vivo and in vitro. Chin J Integr Med. 18:431–436. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Shen AL, Hong F, Liu LY, Lin JM, Wei LH, Cai QY, Hong ZF and Peng J: Pien Tze Huang inhibits the proliferation of human colon carcinoma cells by arresting G1/S cell cycle progression. Oncol Lett. 4:767–770. 2012.PubMed/NCBI

37 

Shen AL, Chen YQ, Hong F, Lin JM, Wei LH, Hong ZF, Sferra TJ and Peng J: Pien Tze Huang suppresses IL-6-inducible STAT3 activation in human colon carcinoma cells through induction of SOCS3. Oncol Rep. 28:2125–2130. 2012.PubMed/NCBI

38 

Shen A, Lin J, Chen Y, Lin W, Liu L, Hong Z, Sferra TJ and Peng J: Pien Tze Huang inhibits tumor angiogenesis in a mouse model of colorectal cancer via suppression of multiple cellular pathways. Oncol Rep. 30:1701–1706. 2013.PubMed/NCBI

39 

Chen HW, Shen AL, Zhang YC, Chen YQ, Lin JM, Lin W, Sferra TJ and Peng J: Pien Tze Huang inhibits hypoxia-induced epithelial-mesenchymal transition in human colon carcinoma cells via suppression of HIF-1 pathway. Exp Ther Med. 7:1237–1242. 2014.PubMed/NCBI

40 

Wei LH, Chen PY, Chen YQ, Shen AL, Chen HW, Lin W, Hong ZF, Sferra T and Peng J: Pien Tze Huang suppresses the stem-like side population in colorectal cancer cells. Mol Med Rep. 9:261–266. 2014.

41 

Van Cutsem E and Costa F: Progress in the adjuvant treatment of colon cancer: Has it influenced clinical practice? JAMA. 294:2758–2760. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Lippman SM: The dilemma and promise of cancer chemoprevention. Nat Clin Pract Oncol. 10:5232006. View Article : Google Scholar

Related Articles

Journal Cover

February-2015
Volume 46 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shen A, Lin W, Chen Y, Liu L, Chen H, Zhuang Q, Lin J, Sferra TJ and Peng J: Pien Tze Huang inhibits metastasis of human colorectal carcinoma cells via modulation of TGF-β1/ZEB/miR‑200 signaling network. Int J Oncol 46: 685-690, 2015
APA
Shen, A., Lin, W., Chen, Y., Liu, L., Chen, H., Zhuang, Q. ... Peng, J. (2015). Pien Tze Huang inhibits metastasis of human colorectal carcinoma cells via modulation of TGF-β1/ZEB/miR‑200 signaling network. International Journal of Oncology, 46, 685-690. https://doi.org/10.3892/ijo.2014.2772
MLA
Shen, A., Lin, W., Chen, Y., Liu, L., Chen, H., Zhuang, Q., Lin, J., Sferra, T. J., Peng, J."Pien Tze Huang inhibits metastasis of human colorectal carcinoma cells via modulation of TGF-β1/ZEB/miR‑200 signaling network". International Journal of Oncology 46.2 (2015): 685-690.
Chicago
Shen, A., Lin, W., Chen, Y., Liu, L., Chen, H., Zhuang, Q., Lin, J., Sferra, T. J., Peng, J."Pien Tze Huang inhibits metastasis of human colorectal carcinoma cells via modulation of TGF-β1/ZEB/miR‑200 signaling network". International Journal of Oncology 46, no. 2 (2015): 685-690. https://doi.org/10.3892/ijo.2014.2772