Scutellaria barbata D. Don inhibits migration and invasion of colorectal cancer cells via suppression of PI3K/AKT and TGF‑β/Smad signaling pathways

  • Authors:
    • Yiyi Jin
    • Wujin Chen
    • Hong Yang
    • Zhaokun Yan
    • Zijun Lai
    • Jianyu Feng
    • Jun Peng
    • Jiumao Lin
  • View Affiliations

  • Published online on: October 2, 2017     https://doi.org/10.3892/etm.2017.5242
  • Pages: 5527-5534
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Metastasis is one of the most aberrant behaviors of cancer cells. Patients with cancers, including colorectal cancer (CRC), have a higher risk of tumor recurrence and cancer‑related mortality once metastasis is diagnosed. Existing treatment strategies fail to cure cancer mostly due to the onset of metastasis. Therefore, metastasis remains a challenge in cancer treatment. Some complementary and alternative medical therapies using traditional Chinese medicine have been demonstrated to be clinically effective in cancer treatment. Scutellaria barbata D. Don (SB) is a promising medicinal herb. It was previously reported that the ethanol extract of SB (EESB) is able to promote apoptosis, and inhibit cell proliferation and angiogenesis in human colon cancer cells. However, the anticancer effect of SB and the underlying mechanism require further investigation, particularly its role against metastasis. To further elucidate the antimetastatic effect of SB, MTT and Transwell assays were used in the present study to evaluate the effect of EESB on the proliferation, migration and invasion of the CRC cell line HCT‑8. In addition, western blot analysis was performed to detect the expression of matrix metalloproteinases (MMPs), cadherins and other metastasis‑associated proteins. EESB significantly reduced HCT‑8 cell viability and attenuated the migration and invasion ability of HCT‑8 cells in a dose‑dependent manner. In addition, EESB decreased the expression of MMP‑1, MMP‑2, MMP‑3/10, MMP‑9 and MMP‑13, and proteins in the phosphoinositide 3‑kinase (PI3K)/AKT and transforming growth factor (TGF)‑β/Smad pathways, but not the epithelial‑mesenchymal transition (EMT)‑related factors E‑cadherin and N‑cadherin. In conclusion, the results suggested that SB inhibits CRC cell metastasis via the suppression of PI3K/AKT and TGF‑β/Smad signaling pathways, which may represent a mechanism by which SB exerts an anticancer effect.

Introduction

Colorectal cancer (CRC) is one of the most common epithelial cancers (1). Epidemiologically, CRC is the third most prevalent cancer worldwide with a high mortality rate in males and females (2). Although CRC management and therapy are performed by screening, surgery, adjuvant irradiation and chemotherapy, CRC remains one of the most life-threatening malignancies, particularly when it reaches the advanced stages (3,4). Approximately 50% of patients with CRC develop metastasis, which is usually incurable and fatal (5). The majority of patients with CRC and distant metastasis are not suitable candidates for conventional intervention and exhibit a poor 5-year survival rate of <10% (6). From a therapeutic perspective, the identification of molecular mechanisms underlying the metastatic progression of CRC may contribute to the reduction of morbidity and mortality (7). In addition, the discovery of effective and safe compounds for the treatment of CRC is urgently required in order to reduce morbidity and mortality rates.

Tumor metastasis is a complex process, and is highly regulated by multiple mechanisms, including aberrant activation of the phosphoinositide 3-kinase (PI3K)/AKT and transforming growth factor (TGF)-β/Smad pathways (811). Furthermore, a large number of studies have shown that matrix metalloproteinase (MMP) overexpression is involved in numerous malignant tumors, including esophageal cancer, breast cancer, liver cancer and rectal cancer (1216). MMPs serve a very important role in tumor invasion and metastasis (12). In addition, epithelial-mesenchymal transition (EMT) is closely associated with tumor occurrence and metastasis (12,17). Furthermore, the expression of MMP family-related factors or N-cadherin/E-cadherin is regulated by the aforementioned pathways in an interactive manner (18,19). As a result, targeting PI3K/AKT and TGF-β/Smad pathways may represent a novel therapeutic method to prevent metastasis without causing side effects.

Traditional Chinese medicine (TCM) is of interest to researchers as it induces relatively few side effects and has been clinically used for thousands of years as an important alternative remedy for a variety of diseases (2023). TCMs are considered to be multi-component and multi-targeted agents that exert their therapeutic functions holistically (24). Scutellaria barbata D. Don (SB) is a medicinal herb widely distributed in northeast Asia. In TCM, SB is a well-known herb considered to be useful for heat-clearing, detoxification, promotion of blood circulation and removal of blood stasis (25). SB has long been used as an important component in several TCM formulas for the clinical treatment of various types of cancer. SB extracts have been shown to inhibit the growth of numerous cancer cell types (2630). In a previous study, it was reported that SB promotes cancer cell apoptosis via activation of the mitochondrial-dependent pathway (31). However, studies in which the anticancer effect of SB and its mechanisms are elucidated, particularly studies relating to metastasis, are lacking. In the present study, the effects of SB on the migration and invasion abilities of HCT-8 human colorectal carcinoma cells and their regulation through PI3K/AKT and TGF-β/Smad signaling pathways were evaluated.

Materials and methods

Materials and reagents

RPMI-1640 medium, fetal bovine serum (FBS), penicillin-streptomycin and trypsin-EDTA were obtained from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Rabbit polyclonal antibodies against AKT (cat. no. 10176-2-AP) and PI3K (cat. no. 13329-1-AP) were purchased from Proteintech Group (Wuhan, China). Rabbit polyclonal antibodies against phospho (p)-AKT (cat. no. sc-135650) and p-PI3K (cat. no. sc-12929), and goat polyclonal antibodies against phosphatase and tensin homolog (PTEN) (cat. no. sc-6818) were purchased from Santa Cruz Biotechnology (Shanghai) Co., Ltd. (Shanghai, China). Rabbit polyclonal antibodies against MMP1 (cat. no. D120093), 2 (cat. no. D161446), 9 (cat. no. D120097) and 13 (cat. no. D120098)] were purchased from Sangon Biotech Co., Ltd. (Shanghai, China), and MMP3/10 (cat. no. sc-30070) was purchased from Santa Cruz Biotechnology (Shanghai) Co., Ltd. Mouse monoclonal antibodies against E-cadherin (cat. no. ab76055) and N-cadherin (cat. no. ab98952) were purchased from Abcam (Hong Kong) Ltd. (Hong Kong, China). Rabbit polyclonal antibodies against TGF-β1 (cat. no. 3711S), Smad4 (cat. no. 38454S), Smad2/3 (cat. no. 8685S) and β-actin (cat. no. 4967), and horseradish peroxidase (HRP)-conjugated secondary antibodies (cat. no. 7074) were provided by Cell Signaling Technology, Inc. (Beverly, MA, USA). Transwell chambers were obtained from Corning Life Sciences (Tewksbury, MA, USA). BD BioCoat Matrigel Invasion Chamber was purchased from BD Biosciences (San Jose, CA, USA). All other chemicals were obtained from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany) unless otherwise stated.

Ethanol extract of SB (EESB) preparation

Authentic plant material was purchased from Guo Yi Tang Chinese herbal medicine store (Fujian, China). The original herb was identified as SB by Dr Wei Xu at the Department of Pharmacology, Fujian University of Traditional Chinese Medicine (Fuzhou, China). The plants were dried and cut into small pieces, and EESB was obtained as previously described (32). EESB stock solutions were prepared by dissolving EESB powder in PBS at a concentration of 500 mg/ml, and stored at −20°C. EESB working concentrations were obtained by diluting the stock solution in the culture medium.

Cell culture

HCT-8 human colorectal carcinoma cells were obtained from Nanjing KeyGen Biotech. Co., Ltd. (Nanjing, 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. Cells were digested at room temperature for 3 min using trypsin-EDTA and subcultured when 80–90% confluency was reached.

MTT assay

Cell viability was assessed using an MTT colorimetric assay. Cells were harvested, re-suspended at a final concentration of 1×105 cells/ml and then seeded into 96-well plates at a volume of 100 µl/well. After 12 h incubation at 37°C, cells were treated with EESB at different concentrations (0, 0.125, 0.25, 0.5, 1, 1.5 and 2 mg/ml) and incubated for 24 or 48 h. Subsequently, 100 µl MTT (0.5 mg/ml) was added to each well. The plates were incubated at 37°C for 4 h, and 100 µl DMSO was added to dissolve the purple formazan crystals. The absorbance was read at 570 nm using an ELISA reader (Model ELx800; BioTek Instruments, Inc., Winooski, VT, USA).

Microscopic observation of cell density

HCT-8 cells were seeded into 6-well plates at a density of 5×105 cells/well in 2 ml medium. Cells were treated with EESB at different concentrations (0, 0.125, 0.25 and 0.5 mg/ml) and incubated for 24 h. Cell density was observed using a phase-contrast microscope (Leica Microsystems GmbH, Wetzlar, Germany). Images were captured at a magnification of ×200.

Cell migration and invasion analysis using Transwell assays

Migration assays were performed using Transwell cell culture chambers with 8-µm pore filters (Corning Life Sciences). Following treatment with EESB at different concentrations (0, 0.125, 0.25 and 0.5 mg/ml) for 24 h, HCT-8 cells were trypsinized and resuspended in serum-free RPMI-1640. A total of 5×104 cells in 200 µl serum-free RPMI-1640 were plated in the upper chamber. RPMI-1640 media containing 10% (v/v) FBS was placed in the lower chamber as a chemoattractant. Cells were allowed to migrate for 12 h, and the non-migrated cells were removed from the upper surface of the Transwell membranes using a cotton swab. Membranes were fixed with ice-cold 4% paraformaldehyde for 10 min and stained using crystal violet at room temperature for 15 min. The average number of migrating cells per field was assessed by counting three random fields under a phase-contrast microscope (Leica) at a magnification of ×200. The procedure for the cell invasion assay was the same as that described for the migration assay, with the exception that the upper chamber was coated with Matrigel Matrix (BD Biosciences).

Western blot analysis

HCT-8 cells were seeded into 25-cm2 flasks at a density of 1.25×106 cells/flask in 5 ml medium. Following incubation for 12 h, cells were treated with EESB at different concentrations (0, 0.125, 0.25 and 0.5 mg/ml) and incubated for 24 h. The treated cells were lysed using Pierce radioimmunoprecipitation assay buffer (Thermo Fisher Scientific, Inc.) containing EASYpack protease and PhosSTOP phosphatase inhibitor cocktails (both Roche Diagnostics, Basel, Switzerland). The lysates were then centrifuged at 17,000 × g for 20 min at 4°C, and the total protein concentration was determined by BCA assay. Equal amounts of total proteins (50 mg) were resolved using 10% SDS-PAGE gels and then electroblotted onto nitrocellulose membranes. The membranes were blocked with 5% nonfat dry milk at room temperature for 2 h, and treated with primary antibodies against E-cadherin (1:1,000), N-cadherin (1:1,000), TGF-β1 (1:1,000), Smad2/3 (1:1,000), Smad4 (1:1,000), AKT (1:500), p-AKT (1:500), PTEN (1:500), PI3K (1:500), p-PI3K (1:500), MMP1 (1:1,000), MMP2 (1:1,000), MMP3/10 (1:1,000), MMP9 (1:1,000), MMP13 (1:1,000) and β-actin (1:1,000) overnight at 4°C. Subsequently, the membranes were incubated with HRP-conjugated secondary antibody at room temperature for 1 h and the protein bands were detected using an enhanced chemiluminescence detection reagent, SuperSignal West Pico Chemiluminescent substrate (Thermo Fisher Scientific, Inc.). β-actin was used as the internal control. Images were taken using a ChemiDoc XRS+ imaging system (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Image Lab™ software (version 3.0; Bio-Rad Laboratories, Inc.) was used for densitometric analysis and quantification of the western blots.

Statistical analysis

All data were obtained as the mean of three experiments. Statistical analysis was performed using SPSS software (version 17.0) for Windows (SPSS, Inc., Chicago, IL, USA) using one-way analysis of variance, followed by Fisher's least significant difference and Dunnett's tests. Data are expressed as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of EESB on HCT-8 cell viability

As shown in Fig. 1, EESB at low concentrations (0.125, 0.25 and 0.5 mg/ml) did not exhibit a significant effect on HCT-8 cell proliferation, while EESB at high concentrations (1, 1.5 and 2 mg/ml) significantly inhibited HCT-8 cell growth compared with that of the untreated cells. On the basis of these results, EESB concentrations of 0.125, 0.25 and 0.5 mg/ml were selected for the subsequent experiments. To further verify that the selected concentrations of EESB were not cytotoxic, the effect of EESB on HCT-8 cell density was analyzed under a microscope. As shown in Fig. 2, as the drug concentration increased from 0 to 0.5 mg/ml, there was no clear change in cell density, which indicated that these low doses of EESB had no marked effect on cell growth.

Effect of EESB on HCT-8 cell migration and invasion

As shown in Fig. 3A and B, EESB significantly reduced the number of migrated cells compared with the untreated control. Similarly, EESB treatment significantly reduced cell invasion through the Matrigel membrane compared with the untreated control (Fig. 3C and D). The inhibitory effects on migration and invasion were concentration-dependent. The number of migrated cells was reduced by 20.89% using 0.125 mg/ml EESB and by 99.89% using 0.5 mg/ml EESB, as compared with the untreated control cells (Fig. 3B). The invasion assay results showed that EESB treatment for 24 h reduced the HCT-8 cell invasion rate by 11.86% when 0.125 mg/ml EESB was used and by 66.90% when 0.5 mg/ml EESB was used, as compared with the untreated control cells (Fig. 3D).

Effect of EESB on MMP and E-/N-cadherin expression

To elucidate the antimetastatic mechanisms of EESB, the expression levels of MMPs (MMP1, MMP2, MMP3/10, MMP9 and MMP13) and the EMT-regulated factors (E-cadherin and N-cadherin) were analyzed using western blotting. As shown in Fig. 4, EESB significantly inhibited the expression of MMP1, MMP2, MMP3/10, MMP9 and MMP13 to different extents, but exerted only a slight effect on the expression of the mesenchymal marker N-cadherin and the epithelial marker E-cadherin. These results indicated that EESB inhibited HCT-8 metastasis via the suppression of MMP expression but not via EMT.

Effect of EESB on PI3K/AKT and TGF-β/Smad signaling pathways

EESB markedly suppressed the activation of the PI3K/AKT pathway by significantly downregulating p-PI3K, PI3K and p-AKT protein levels (Fig. 5). In addition, PTEN, which is a tumor suppressor and PI3K/AKT upstream factor (33), was significantly upregulated following EESB treatment. Furthermore, EESB treatment significantly inhibited the expression of TGF-β, Smad2/3 and Smad4. These results suggested that the antimetastatic effect of EESB on CRC may be partly mediated by suppression of the PI3K/AKT and TGF-β/Smad signaling pathways.

Discussion

CRC remains a potentially lethal disease with a poor prognosis, mostly due to metastasis in the majority of patients. Multi-drug combination therapies have been developed leading to significantly improved patient response and overall survival. However, resistance to these drugs is inevitable and continues to be a notable problem (34,35). Therefore, novel agents, including natural products, are currently being considered for more efficient cancer treatment. TCM, with its relatively high safety and long history of pharmacological applications, has attracted attention in the field of cancer treatment (20,23). SB is a herb used in TCM formulations, where it is considered to have ‘heat-clearing and detoxifying’ actions, and has many reported applications in cancer treatment (3641). Similar to other medicinal herbs, SB is a multi-targeted agent that is considered to exert its therapeutic function holistically (27,31,32,42); thus, it may be a good candidate as an anticancer drug. However, the specific mechanism of its anticancer effect, particularly its antimetastatic ability, is not yet clear.

The process of tumor metastasis is complex. One of the most studied mechanisms relates to MMP involvement. MMPs are a group of important proteases that degrade extracellular matrix (ECM). Numerous studies have shown that MMPs are associated with tumor growth, metastasis and invasion (4345). The main members of the family may be divided into five groups according to their domain, enzyme and substrate specificity as follows: Collagenase (MMP1, MMP8 and MMP13), gelatinase (MMP2 and MMP9), matrix soluble elements (MMP3, MMP7, MMP10 and MMP11) and matrix dissolution factor and membrane type (MT) metalloproteinase (MT1-MMP, MT2-MMP and MT3-MMP) (46). It has been reported that MMP1 expression is increased significantly in gastric cancer, which destroys the basement membrane, and promotes tumor lymphangiogenesis, tumor invasion and metastasis (47). MMP2 has been demonstrated to be closely associated with migration and invasion in several types of tumors, including breast cancer, ovarian cancer and lung cancer (48,49). MMP9 expression was identified to be increased in osteosarcoma, lung cancer, pancreatic cancer and CRC tissues to different extents, which was positively correlated with tumor metastasis (4850). Similarly, it has been observed that MMP3 and MMP10 expression levels in lung cancer, esophageal cancer, liver cancer and endometrial adenocarcinoma tissues are higher than those in normal tissues, and that their expression has an association with tumor invasion, metastasis and proliferation (5155). In addition, a review of a number of studies has demonstrated that MMP13 expression in malignant tumors, such as colon cancer, breast cancer, non-small cell lung cancer and oral squamous cell carcinoma, is closely associated with tumor occurrence, development, invasion and metastasis (56). On the basis of this evidence, MMPs have a close association with tumor invasion and metastasis, serving a very important role. In the present study, EESB downregulated the expression of the MMPs, to different extents, suggesting that EESB may inhibit CRC metastasis by inhibiting the expression of these specific MMPs to balance the ECM environment.

EMT is an important phenomenon in the occurrence and development of tumors and is also an important mechanism allowing tumor invasion and metastasis (57,58). E-cadherin and N-cadherin are two important factors in the maintenance of the EMT balance; EMT regulation is influenced by these two cadherins (17). However, in the present study, EESB showed only a weak effect on the expression of the mesenchymal marker N-cadherin and epithelial marker E-cadherin, suggesting that EESB inhibits CRC metastasis thorough mechanisms other than EMT.

In addition to the aforementioned factors, numerous pathways also contribute to tumor metastasis. For example, activation of the PI3K/AKT signaling pathway accelerates angiogenesis, tumor invasion and metastasis through the disturbance of tumor-suppressor PTEN or other causal factors, which is important in the occurrence and development of malignant tumors (59). Furthermore, the PI3K/AKT signaling pathway is involved in regulating the expression of MMP-2 and MMP-9 in a variety of tumor tissues and cells, to regulate multidrug resistance, as well as tumor invasion and metastasis (60). Furthermore, TGF-β promotes tumor metastasis by increasing tumor angiogenesis, immune suppression, and the production and deposition of ECM (61). Previous studies have shown that TGF-β may cause tumor metastasis through activation of the Smad pathway, and Smad2 and Smad4 are important proteins that regulate the transcription and the antiproliferative response of TGF-β, and regulate the expression of genes downstream of TGF-β that are involved in tumor metastasis (62,63). Furthermore, TGF-β may mediate tumor cell invasion by regulating ECM-degrading proteinases (64). Among the increasing number of ECM-degrading proteinases implicated in tumor cell invasion, the majority of the attention has been focused on the MMP family and the plasminogen activator system (65,66). TGF-β1 has been suggested to activate the Smad signaling pathway, and significantly promote the expression of MMPs and other invasion and metastasis-related genes in highly invasive breast cancer cells, thereby enhancing the ability of the cells to invade and metastasize (67,68). In the present study, EESB decreased the expression of proteins in the PI3K/AKT and TGF-β/Smad2/3 pathways, and upregulated the expression of the tumor-suppressor PTEN, thus indicating that the inhibitory effect of EESB on metastatic CRC cells may be mediated by the suppression of certain members of the MMP family, and PI3K/AKT and TGF-β/Smad signaling pathways.

In conclusion, EESB exerted significant antimetastatic effects on CRC cells by inhibition of their migration and invasion ability, and via the regulation of PI3K/AKT and TGF-β/Smad signaling pathways. These mechanisms are potentially those by which EESB exhibits its effectiveness in cancer treatment.

Acknowledgements

The present study was sponsored by the Research Fund for the Doctoral Program of Higher Education of China (grant no. 20133519110003), the Project Funding for the Training of Young and Middle-aged Backbone Personnel of Fujian Provincial Health and Family Planning Commission (grant no. 2016-ZQN-67), and the Developmental Fund of Chen Keji Integrative Medicine (Fujian, China; grant nos. CKJ2014013 and CKJ2015007).

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

EESB

ethanol extract of Scutellaria barbata D. Don

TCM

traditional Chinese medicine

ECM

extracellular matrix

MMP

matrix metalloproteinase

EMT

epithelial-mesenchymal transition

References

1 

Tenesa A and Dunlop MG: New insights into the aetiology of colorectal cancer from genome-wide association studies. Nat Rev Genet. 10:353–358. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Grávalos C, Cassinello J, Fernández-Rañada I and Holgado E: Role of tyrosine kinase inhibitors in the treatment of advanced colorectal cancer. Clin Colorectal Cancer. 6:691–699. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Brenner H, Kloor M and Pox CP: Colorectal cancer. Lancet. 383:1490–1502. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Van Cutsem E and Oliveira J: ESMO Guidelines Working Group: Advanced colorectal cancer: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 20 Suppl 4:S61–S63. 2009. View Article : Google Scholar

6 

Manfredi S, Lepage C, Hatem C, Coatmeur O, Faivre J and Bouvier AM: Epidemiology and management of liver metastases from colorectal cancer. Ann Surg. 244:254–259. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Talmadge JE and Fidler IJ: AACR centennial series: The biology of cancer metastasis: Historical perspective. Cancer Res. 70:5649–5669. 2010. View Article : Google Scholar : PubMed/NCBI

8 

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. View Article : Google Scholar : PubMed/NCBI

9 

Kalluri R and Weinberg RA: The basics of epithelial-mesenchymal transition. J Clin Invest. 119:1420–1428. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Xu T, Jing C, Shi Y, Miao R, Peng L, Kong S, Ma Y and Li L: microRNA-20a enhances the epithelial-to-mesenchymal transition of colorectal cancer cells by modulating matrix metalloproteinases. Exp Ther Med. 10:683–688. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Alam SK, Yadav VK, Bajaj S, Datta A, Dutta SK, Bhattacharyya M, Bhattacharya S, Debnath S, Roy S, Boardman LA, et al: DNA damage-induced ephrin-B2 reverse signaling promotes chemoresistance and drives EMT in colorectal carcinoma harboring mutant p53. Cell Death Differ. 23:707–722. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Guo S: Research progress in the mechanism of colorectal cancer metastasis. J Mudanjiang Med College. 29:65–67. 2008.

13 

Juchniewicz A, Kowalczuk O, Milewski R, Laudański W, Dzięgielewski P, Kozłowski M and Nikliński J: MMP-10, MMP-7, TIMP-1 and TIMP-2 mRNA expression in esophageal cancer. Acta Biochim Pol. 64:295–299. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Yun EJ, Song KS, Shin S, Kim S, Heo JY, Kweon GR, Wu T, Park JI and Lim K: Docosahexaenoic acid suppresses breast cancer cell metastasis by targeting matrix-metalloproteinases. Oncotarget. 7:49961–49971. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Dou CY, Cao CJ, Wang Z, Zhang RH, Huang LL, Lian JY, Xie WL and Wang LT: EFEMP1 inhibits migration of hepatocellular carcinoma by regulating MMP2 and MMP9 via ERK1/2 activity. Oncol Rep. 35:3489–3495. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Fuksiewicz M, Kotowicz B, Rutkowski A and Kowalska M: The matrix metalloproteinase-7 and pro-enzyme of metalloproteinase-1 as a potential marker for patients with rectal cancer without distant metastasis. Tumour Biol. 36:3629–3635. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Zhang M, Zhuo N and Guo Z: Molecular mechanism of epithelial-mesenchymal transition and its role in tumor metastasis. Chin Med Herald. 11:163–165. 2014.

18 

Wei F, Shen Q and Liu C: Phenethyl isothiocyanate inhibits PI3K/NF-kB to down-regulate MMP-9 expression in human colon cancer cells. Med Sci J Central South China. 42:351–354. 2014.

19 

Liang S, Lv Y and Wang X: Study of the correlation and expression of focal adhesion kinase and matrix metalloproteinase-9 in colorectal carcinoma. J Colorectal Anal Surger. 14:157–160. 2008.

20 

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

21 

Zhao J, Jiang P and Zhang W: Molecular networks for the study of TCM pharmacology. Brief Bioinform. 11:417–430. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Lin J, Chen Y, Wei L, Chen X, Xu W, Hong Z, Sferra TJ and Peng J: Hedyotis Diffusa Willd extract induces apoptosis via activation of the mitochondrion-dependent pathway in human colon carcinoma cells. Int J Oncol. 37:1331–1338. 2010.PubMed/NCBI

23 

Demain AL and Zhang L: Natural Products and Drug Discovery. Can thousands of years of ancient medical knowledge lead us to new and powerful drug combinations in the fight against cancer and dementia? EMBO Rep. 10:194–200. 2009. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Read BE: The Chinese Pharmacopoeia. Can Med Assoc J. 23:568–570. 1930.PubMed/NCBI

26 

Cha YY, Lee EO, Lee HJ, Park YD, Ko SG, Kim DH, Kim HM, Kang IC and Kim SH: Methylene chloride fraction of Scutellaria barbata induces apoptosis in human U937 leukemia cells via the mitochondrial signaling pathway. Clin Chim Acta. 348:41–48. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Lin J, Chen Y, Cai Q, Wei L, Zhan Y, Shen A, Sferra TJ and Peng J: Scutellaria barbata D Don inhibits colorectal cancer growth via suppression of multiple signaling pathways. Integr Cancer Ther. 13:240–248. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Suh SJ, Yoon JW, Lee TK, Jin UH, Kim SL, Kim MS, Kwon DY, Lee YC and Kim CH: Chemoprevention of Scutellaria bardata on human cancer cells and tumorigenesis in skin cancer. Phytother Res. 21:135–141. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Yin X, Zhou J, Jie C, Xing D and Zhang Y: Anticancer activity and mechanism of Scutellaria barbata extract on human lung cancer cell line A549. Life Sci. 75:2233–2244. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Lee TK, Lee DK, Kim DI, Lee YC, Chang YC and Kim CH: Inhibitory effects of Scutellaria barbata D. Don on human uterine leiomyomal smooth muscle cell proliferation through cell cycle analysis. Int Immunopharmacol. 4:447–454. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Wei L, Chen Y, Lin J, Zhao J, Chen X, Xu W, Liu X, Sferra T and Peng J: Scutellaria barbata D. Don induces apoptosis of human colon carcinoma cell through activation of the mitochondrion-dependent pathway. J Med Plants Res. 5:1962–1970. 2011.

32 

Wei L, Lin J, Wu G, Xu W, Li H, Hong Z and Peng J: Scutellaria barbata D. Don induces G1/S arrest via modulation of p53 and Akt pathways in human colon carcinoma cells. Oncol Rep. 29:1623–1628. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Georgescu MM: PTEN tumor suppressor network in PI3K-Akt pathway control. Genes Cancer. 1:1170–1177. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Rodel C, Hofheinz R and Liersch T: Rectal cancer: State of the art in 2012. Curr Opin Oncol. 24:441–447. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Cunningham D, Atkin W, Lenz HJ, Lynch HT, Minsky B, Nordlinger B and Starling N: Colorectal cancer. Lancet. 375:1030–1047. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Tan P, Lu B and Bao W: Analysis on the clinical application of Scutellaria barbata D. Don in the anti-cancer therapy. Jiangxi Tradit China Med. 37:57–58. 2006.

37 

Dai ZJ, Liu XX, Tang W, Xue Q, Wang XJ, Ji ZZ, Kang HF and Diao Y: Antitumor and immune-modulating effects of Scutellaria barbata extract in mice bearing hepatocarcinoma H22 cells-derived tumor. Nan Fang Yi Ke Da Xue Xue Bao. 28:1835–1837. 2008.(In Chinese). PubMed/NCBI

38 

Goh D, Lee YH and Ong ES: Inhibitory effects of a chemically standardized extract from Scutellaria barbata in human colon cancer cell lines, LoVo. J Agric Food Chem. 53:8197–8204. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Marconett CN, Morgenstern TJ, San Roman AK, Sundar SN, Singhal AK and Firestone GL: BZL101, a phytochemical extract from the Scutellaria barbata plant, disrupts proliferation of human breast and prostate cancer cells through distinct mechanisms dependent on the cancer cell phenotype. Cancer Biol Ther. 10:397–405. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Wong BY, Nguyen DL, Lin T, Wong HH, Cavalcante A, Greenberg NM, Hausted RP and Zheng J: Chinese medicinal herb Scutellaria barbata modulates apoptosis and cell survival in murine and human prostate cancer cells and tumor development in TRAMP mice. Eur J Cancer Prev. 18:331–341. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Zhao Z, Holle L, Song W, Wei Y, Wagner TE and Yu X: Antitumor and anti-angiogenic activities of Scutellaria barbata extracts in vitro are partially mediated by inhibition of Akt/protein kinase B. Mol Med Rep. 5:788–792. 2012.PubMed/NCBI

42 

Zhang L, Cai Q, Lin J, Fang Y, Zhan Y, Shen A, Wei L, Wang L and Peng J: Chloroform fraction of Scutellaria barbata D. Don promotes apoptosis and suppresses proliferation in human colon cancer cells. Mol Med Rep. 9:701–706. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Herszényi L, Hritz I, Lakatos G, Varga MZ and Tulassay Z: The behavior of matrix metalloproteinases and their inhibitors in colorectal cancer. Int J Mol Sci. 13:13240–13263. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Littlepage LE, Sternlicht MD, Rougier N, Phillips J, Gallo E, Yu Y, Williams K, Brenot A, Gordon JI and Werb Z: Matrix metalloproteinases contribute distinct roles in neuroendocrine prostate carcinogenesis, metastasis, and angiogenesis progression. Cancer Res. 70:2224–2234. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Laurie AS, Sandra JT and William GS: Matrix metalloproteinases: Changing roles in tumor progression and metastasis. Am J Pathol. 181:1895–1899. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Gao Y and Wang S: Relationship between matrix metalloproteinases and their inhibitors and their relationship with invasion and metastasis of malignant tumors. J New Med. 42:341–343. 2011.

47 

Liu T, Ma Y and Zhang R: Advance research on relationship between matrix metalloproteinases and invasion and metastasis of malignant tumors. J Jilin Univ. 30:662–664. 2004.

48 

Meng F, Liu X and Qi S: Correlation of matrix metalloproteinases-2 and-9 in ovarian cancer. Chin J Gerontology. 33:3505–3506. 2013.

49 

Ming S, Sun T and Xiao W: Role of matrix metalloproteinases −2, −9 and its inhibitor 1 in the invasion and metastasis of lung cancer. Chin J Respirator Crit Care. 4:198–202. 2005.

50 

Zheng H, Shen B, Nie Y and Du Y: The expression of MMP-9, MMP-13 and TIMP-3 in hepatocellular carcinoma. Guangdong Med J. 34:1995–1998. 2013.

51 

Yan Z, Xu X and Yang G: Abnormal expression and clinical significance of matrix metalloprteinase 10 in esophagus carcinoma. Ningxia Med J. 27:14–15. 2005.

52 

He J, Ding C, He G and Huang Q: Relationship between expression of ESM-1 and MMP-3 and invasion and metastasis of human hepatocellular carcinoma. Med Sci J Central South China. 40:368–372. 2012.

53 

Yue X, Zhang Q, Xu A, Xing Y and Zhang F: Expression and clinical significance of MMP10 and CD105 in patients with non-small-cell lung carcinoma. Shandong Med J. 49:13–15. 2009.

54 

Feng J, Gou W, Liu D and Li X: Expressions of matrix metalloproteinase-3 and matrix metalloproteinase-10 in endometrial carcinoma. J Xian Jiaotong Univ (Med Sci). 31:97–101. 2010.

55 

Tian J, Xu M and Jing H: Matrix metalloproteinases 3 and its inhibitory factor 2 expression in lung squamous carcinoma. West China Med J. 28:369–372. 2013.

56 

Ma R and Zhang C: Review of matrix metalloproteinases-13 and its relationship with invasion and metastasis of malignant tumor. J Community Med. 10:47–49. 2012.

57 

Da C, Liu Y, Zhan Y, Liu K and Wang R: Nobiletin inhibits epithelial-mesenchymal transition of human non-small cell lung cancer cells by antagonizing the TGF-β1/Smad3 signaling pathway. Oncol Rep. 35:2767–2774. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Ji Q, Liu X, Han Z, Zhou L, Sui H, Yan L, Jiang H, Ren J, Cai J and Li Q: Resveratrol suppresses epithelial-to-mesenchymal transition in colorectal cancer through TGF-β1/Smads signaling pathway mediated Snail/E-cadherin expression. BMC Cancer. 15:972015. View Article : Google Scholar : PubMed/NCBI

59 

Guo L and Wang Q: Correlation of PI3K/AKT/mTOR signal pathway to infiltration and metastasis of malignant tumor. Modern Oncol. 17:1585–1589. 2009.

60 

Guo C, Ke W, Song K, Wang J, Zhou L and Li K: PI3K/AKT signalling pathway involves in the modulation of multidrug resistance and metastasis in breast cancer. Prog Mod Biomed. 12:4809–4812. 2012.

61 

Jia BY and Wang YJ: Metastasis associated signaling pathway in colorectal cancer. Int J Dig Dis. 3:183–185. 2015.

62 

Ding P, Song B and Zhu L: Effect of TGF-β/Smad 4 on the tumor metastasis of colorectal cancer cells. Chin J Cancer Prev Treat. 18:1518–1520. 2011.

63 

Qi Y and Li H: Research of Qilian Fuzheng capsule's function on anti-lung cancer metastasis by regulating TGF-β pathway. Chin Archives Tradit Chin Med. 32:2567–2569. 2014.

64 

Liotta LA: Tumor invasion and metastases-role of the extracellular matrix. Rhoads Memorial Award lecture. Cancer Res. 46:1–7. 1986.PubMed/NCBI

65 

Stetler-Stevenson WG, Hewitt R and Corcoran M: Matrix metalloproteinases and tumor invasion: From correlation and causality to the clinic. Semin Cancer Biol. 7:147–154. 1996. View Article : Google Scholar : PubMed/NCBI

66 

Chambers AF and Matrisian LM: Changing views of the role of matrix metalloproteinases in metastasis. J Natl Cancer Inst. 89:1260–1270. 1997. View Article : Google Scholar : PubMed/NCBI

67 

Perera M, Tsang CS, Distel RJ, Lacy JN, Ohno-Machado L, Ricchiuti V, Samaranayake LP, Smejkal GB, Smith MG, Trachtenberg AJ and Kuo WP: TGF-beta1 interactome: Metastasis and beyond. Cancer Genomics Proteomics. 7:217–229. 2010.PubMed/NCBI

68 

Roberts AB and Wakefield LM: The two faces of transforming growth factor beta in carcinogenesis. Proc Natl Acad Sci USA. 100:pp. 8621–8623. 2003, View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 14 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jin Y, Chen W, Yang H, Yan Z, Lai Z, Feng J, Peng J and Lin J: Scutellaria barbata D. Don inhibits migration and invasion of colorectal cancer cells via suppression of PI3K/AKT and TGF‑β/Smad signaling pathways. Exp Ther Med 14: 5527-5534, 2017
APA
Jin, Y., Chen, W., Yang, H., Yan, Z., Lai, Z., Feng, J. ... Lin, J. (2017). Scutellaria barbata D. Don inhibits migration and invasion of colorectal cancer cells via suppression of PI3K/AKT and TGF‑β/Smad signaling pathways. Experimental and Therapeutic Medicine, 14, 5527-5534. https://doi.org/10.3892/etm.2017.5242
MLA
Jin, Y., Chen, W., Yang, H., Yan, Z., Lai, Z., Feng, J., Peng, J., Lin, J."Scutellaria barbata D. Don inhibits migration and invasion of colorectal cancer cells via suppression of PI3K/AKT and TGF‑β/Smad signaling pathways". Experimental and Therapeutic Medicine 14.6 (2017): 5527-5534.
Chicago
Jin, Y., Chen, W., Yang, H., Yan, Z., Lai, Z., Feng, J., Peng, J., Lin, J."Scutellaria barbata D. Don inhibits migration and invasion of colorectal cancer cells via suppression of PI3K/AKT and TGF‑β/Smad signaling pathways". Experimental and Therapeutic Medicine 14, no. 6 (2017): 5527-5534. https://doi.org/10.3892/etm.2017.5242