Anti-proliferation effects of trifolirhizin on MKN45 cells and possible mechanism

  • Authors:
    • Xingang Lu
    • Jianxia Ma
    • Hongfu Qiu
    • Liu Yang
    • Lei Cao
    • Jie Shen
  • View Affiliations

  • Published online on: September 23, 2016     https://doi.org/10.3892/or.2016.5125
  • Pages: 2785-2792
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Trifolirhizin is a compound isolated from Sophora flavescens. It has been shown to exert cytotoxicity on several cancer cell lines. However, the underlying mechanism remains unknown. MKN45 cells were used as a research model. We assessed the cytotoxicity of trifolirhizin to MKN45 by MTT. Hoechst staining and TUNEL method were used to demonstrate apoptosis. Flow cytometry was used to determine cell cycle and ratio of apoptosis. Caspase activity assay was used to examine the activation of caspase cascade pathways. Western blotting was used to explore the protein levels. Consistently, trifolirhizin inhibited MKN45 xenograft tumor growth in vivo. Trifolirhizin caused a significantly decreased proliferation of MKN45 cells in a time- and dose-dependent manner, with IC50 values of 33.27±2.06 µg/ml at 48 h. Western blot assay manifested that trifolirhizin activated the EGFR-MAPK signaling pathways. This study indicated that trifolirhizin may be a therapeutic application in human gastric cancer therapy.

Introduction

Gastric cancer is one of prevailing cancers worldwide accounting for nearly 8–10% of cancer mortality (1). According to published reports, there are approximately one million gastric cancer cases with 73,800 deaths in 2008 (2). In recent years, many advances have taken place in diagnosis and treatment (3). However, the mortality of gastric cancer remains 70% especially with late stage patients (4). Pharmacotherapy remains the effective strategy for treatment of gastric cancer (5). Screening effective agents from herb plants have been proved efficient (6). Many constituent have been demonstrated with antitumor effects both in vitro and in vivo, for instance: celastrol (7), bigelovin (8), and parthenolide (9). Other natural compounds also demonstrated many functions leading to cell proliferation, cell differentiation, apoptosis and cell cycle changes (10,11).

Sophora flavescens have been widely planted in East Asian countries as herbal plants and cosmetics (12). The root of Sophora flavescens was frequently used for treatment of mental health, inflammation, asthmas, and microbial activities (1315). Trifolirhizin is a natural pterocarcan flavonoid extracted from Sophora flavescens (16). Published reports demonstrated its chemical structure (Fig. 1A) (17). Trifolirhizin has many pharmacological activities, such as hepatoprotective (18), anti-inflammatory (19), anti-proliferation (20), and skin-whitening (21). Trifolirhizin is also a biologically active constituent of Zeng-Sheng-Ping, a commercial Chinese traditional medicine for cancer prevention (22). Accumulating reports manifested that trifolirhizin had anti-proliferative activities in melanoma B16 cells, human A2780 ovarian and H23 lung cancer cells in vitro (19,21). Trifolirhizin also induced apoptosis in human leukemia HL-60 cells in vitro (23). Despite reports on the trifolirhizin cytotoxicity effect on various cells, the intrinsic mechanism of cytotoxicity and apoptosis triggered by trifolirhizin remains unknown. Herein, it is demonstrated that trifolirhizin induce apoptosis and cell cycle arrest while we explored also the mechanism of activation.

Materials and methods

Reagents and trifolirhizin preparation

Trifolirhizin standard preparation (purity >98%) were purchased from Chinese National Institute for the Control of Pharmaceutical and Biological Products (Beijing, China). All other chemicals used were provided from Sigma-Aldrich (St. Louis, MQ, USA). Trifolirhizin was dissolved in DMSO. Cells were treated with increasing concentrations of trifolirhizin for designated hours. Control groups were exposed to the same volumes of DMSO under the same conditions.

Cell culture and cell viability assay

MKN45, L02, HEK293 cells were purchased from the Cell Bank of Chinese Academy of Sciences (Shanghai, China). All cells were maintained in RPMI-1640 medium (Gibco, Carlsbad, CA, USA) with 10% fetal calf serum (Gibco) containing antibiotics (100 IU/ml penicillin and 100 IU/ml streptomycin) at 37°C in an atmosphere containing 5% CO2. The 3 types of cell lines were treated with various concentrations of trifolirhizin for suitable times. Cell viability of control and experiment cells were investigated using the 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazolium bromide (MTT) assay. The absorbance at 570 nm was assessed using a microplate reader (Perkin-Elmer Inc., Waltham, MA, USA).

Apoptosis detection of Hoechst 33342 staining and TUNEL staining

MKN45 cells (105) were cultured on cover-slide and treated as described above. For Hoechst 33342 staining, cells were added with Hoechst 33342 (5 µg/ml) at 4°C for 10 min in the dark (Beyotime, Haimeng, China). For terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) staining (Roche, Basel, Switzerland), TUNEL kit was used to analyze cell DNA fragmentation in apoptotic cells according to the manufacturer's instructions. Trifolirhizin-treated cells after fixing was added by moderate TUNEL and DAPI (Beyotime). Finally, apoptotic cells were observed using an inverted fluorescence microscope (D5100, Nicon, Tokyo, Japan).

Flow cytometry assay for ratio of apoptosis

MKN45 cell (2–5×105) apoptosis quantification was analyzed according to the Annexin V&PI kit manufacturer's instructions (Selleck Chemicals, Houston, TX, USA). A minimum of 105 trifolirhizin-treated cells were counted for each detection and immediately assessed by FACSCalibur flow cytometry (BD Biosciences, San Jose, CA, USA). Final sample data were reported as percentage of apoptotic cells (sum of early-phase apoptosis at right down quadrant and late-apoptosis at right upper quadrant).

Assay of mitochondrial membrane potential (MMP)

The JC-10 assay kit (AAT Bioquest, CA, USA) was used to assess the change tendency in mitochondrial membrane potential (MMP). The kit was applied according to the manufacturer's instructions. MKN45 cells were incubated in 96-well plate and exposed to trifolirhizin under desired concentrations. JC-10 working solutions (50 µl) were added into each well. After incubation and re-suspending in buffer B, the treated and control MKN45 cell suspensions were measured with an excitation filter of 490 nm while emission filter of 525 nm (green) and another excitation filter of 490 nm while emission filter of 590 nm (red) by a microplate reader (Perkin-Elmer).

Cell cycle assay

PI was employed for cycle analysis (Beyotime). After trifolirhizin treatment at 48 h, MKN45 carcinoma cells were harvested from 6-cm culture plates and subsequently fixed with 70% pre-cooled ethanol. After washing and centrifufation, cell pellets were re-suspended in staining solutions containing RNase A (10 µg/ml) with PI (5 µg/ml) under darkness. Cells treated with trifolirhizin and control were incubated for 30 min and then tested with a FACSCalibur cytometer system (BD Biosciences). Cell cycles were measured using ModFit LT3.2 software.

Western blot analysis

Western blotting was used to investigate the levels of apoptotic and related signaling pathway proteins. Concisely, cells treated with trifolirhizin and control were lysed with lysis buffer (Beyotime) and centrifuged. Cell proteins were fractionated by 8–12% SDS-PAGE. The proteins were electro-transferred onto the nitrocellulose membranes, using the BCA kit to adjust protein levels (Beyotime). The proteins levels were assessed using primary antibody, including Bcl-2, Bax, PARP, c-Myc, caspase-3, caspase-9, p-JNK, p-ERK, p-P38, JNK, P38, ERK, P53, β-actin (CST, Beverly, MA, USA), followed by incubation with anti-mouse/rat HRP-labeled secondary antibodies. The β-actin antibody was performed as control. The signals were invested by ECL chemiluminescence (BD Biosciences). The tendency of increasing or decreasing of proteins levels were analyzed comparing to control.

Animals

The animal study was licensed by the Institutional Animal Care and Use Committee of Fudan University and performed strictly accordance with the Declaration of Helsinki and the Guide for the Care and the Use of Laboratory Animals. The 6-week-old BALB/C nude mice were obtained from Shanghai Slac Laboratory Animal Corporation and maintained in a sterile humidified environment with average temperature at 22°C. The mice were divided into 4 groups: 1 control group and 3 treatment groups (N=6) with increasing trifolirhizin. MKN45 cells were harvested, diluted to desired concentrations (5×106) and injected intraperitoneally into the right flank of each mouse. Then, trifolirhizin was injected into the BALB/C nude mice when tumor volume achieved a mean group size of 100 mm3. The saline was injected into control group. All mouse groups were injected every 3 days. BALB-nu mice were sacrificed at 24 h when tumor volume achieved average group size of 1,000 mm3. MKN45 tumor xenografts were evaluated and fixed for further experiments. Tumor volumes were evaluated as (W2 × L)/2 per 3 days using micrometer calipers, and tumor weights were calculated.

Immunohistochemistry

The xenografted MKN45 tumor tissue samples were removed and cut to desired small pieces. samples (5 µm) were fixed and stained by cleaved-caspase-3 and Ki67 antibodies, respectively, for immunohistochemistry. We observed the visual images using an inverted fluorescence microscope (D5100, Nicon). The immunohistochemistry positive cells were counted and analyzed using ImageJ Pro 6.0 (National Institutes of Health).

Statistical analysis

We performed triplicate experiments using independent samples. Results were expressed as mean values ± standard deviation (mean ± SD). Statistical significance of our present results was determined using the one-way ANOVA. All statistical analyses were performed with commercial SPSS software (SPSS19.0, Chicago, IL, USA).

Results

Trifolirhizin exerts anti-proliferation effects in human gastric cancer cells

Published reports have manifested that trifolirhizin is cytotoxic on cancer cells. MTT assay was used to assess the numbers of viable MKN45 cells. Exposure under trifolirhizin results in a concentration- and time-dependent suppression of MKN45 cell viability (Fig. 1B). The trifolirhizin exposure time was 48 h in the present examination. The IC50 value of trifolirhizin on MKN45 cells was 33.27±2.06 µg/ml. We also conducted MTT assay to evaluate the efficacy of trifolirhizin on a human normal liver cell line and human normal kidney cell line in vitro. To our satisfaction, >60% tested cells were viable when treated with increasing trifolirhizin dose ≤250 µg/ml. The apparently higher IC50 value of trifolirhizin on two normal human cell lines exhibited more safety and further implication possibility (Fig. 1C).

Trifolirhizin induces apoptosis in human stomach cancer cells in vitro

MKN45 cells were treated with desired concentrations of trifolirhizin as above at 48 h. Hoechst staining exhibited visual images with cell shrinkage, nuclear fragmentation and chromatin compaction of apoptosis (Fig. 4A). Images indicated that trifolirhizin lead to increasing population of apoptotic cells. Consistently, TUNEL staining showed a significant difference between trifolirhizin treated cells vs. the vehicle control, the ratio of green staining (apoptotic) vs blue staining (normal) arise with increasing trifolirhizin (20–40 µg/ml, shown in Fig. 4B). Annexin V/PI assay was performed to evaluate the apoptotic population quantitation after trifolirhizin exposure. In this study, trifolirhizin significantly triggered apoptosis. The apoptotic population in trifolirhizin (20–40 µg/ml) induced MKN45 cells increased (Fig. 2). The JC-10 assay was used to indicate the mitochondrial outer membrane permeabilization after trifolirhizin treatment in early phase. Fig. 1D demonstrated that red fluorescence (normal cells) was decreased while green fluorescence (apoptotic cells) increased with trifolirhizin treatment (20–40 µg/ml) at 24 h, so the upgrade ratio of green/red fluorescence reflected that the MMP (∆Ψm) in MKN45 after trifolirhizin treatment (20–40 µg/ml) was decreased.

The expression levels of the epidermal growth factor receptor (EGFR) signaling pathways and its downstream mitogen activated protein kinase (MAPK) signaling pathway were shared involving multiple anticancer agent molecular activities. The EGFR levels, containing RAS/RAF/MEK/ERK are downregulated with the increasing trifolirhizin, Fig. 4A indicates that trifolirhizin (20–40 µg/ml) triggered the increase of p38 and elicited no impact on p-JNK. Bcl-2 and Bax family members are the critical regulators in mitochondrial apoptosis pathways. Trifolirhizin leads to disorder of expression ratio of Bcl-2 and Bax. The induction of apoptosis by trifolirhizin was subsequently mediated through activities of c-Myc, caspase-9, and caspase-3.

Trifolirhizin causes cell cycle arrest in human gastric cancer cells in vitro

PI was employed to determine the cell cycle distribution under trifolirhizin exposure. Fig. 3 shows that trifolirhizin exposure (20–40 µg/ml) resulted in increasing accumulation in G2/M phase in MKN45 cells. Furthermore, trifolirhizin exhibited impact on the cell cycle through regulation of cell cycle related proteins. There are an appreciable downregulation of cyclin B as well as Cdc2 in trifolirhizin treatment (20–40 µg/ml). The P53 protein, which is located upstream of cyclin complex and downstream of EGFR signaling pathways, is a key point between signaling pathway and cellular response in cell cycle arrest. Trifolirhizin treatment (20–40 µg/ml) elicited P53 activation (Fig. 5A).

Anti-proliferation effect of trifolirhizin on MKN45 xenograft tumors

Efficacy of trifolirhizin was also confirmed because there were few changes in the mouse body weight in the experiment (Fig. 1E). These results certified the relative safety of trifolirhizin in vivo. After 21 administration days, xenograft tumors treated with trifolirhizin (1–3 mg/kg) were smaller than control treated with saline (Table I and Fig. 1F). The significantly decreasing levels of Ki67-positive cells were found in the study, which hinted the anti-proliferation effect of trifolirhizin (1–3 mg/kg) in vivo. Cleaved-caspase-3 positive cells were increased which reflected the trifolirhizin apoptotic effect (Fig. 4D). These results also manifested the antitumor effects of trifolirhizin.

Table I

The inhibitory effect of trifolirhizin on MKN45 implantation tumor growth in BALB/C-nu mice.

Table I

The inhibitory effect of trifolirhizin on MKN45 implantation tumor growth in BALB/C-nu mice.

Groups (mg/kg)No. of animals survivedBody weights (g)
Tumor weight (g)Inhibition rate (%)
StartEnd
Control620.35±0.6824.6±0.981.42±0.42
1620.88±0.3624.58±0.740.90±0.23a36.37
2620.71±0.3824.28±1.300.61±0.24b57.29
3620.73±0.4223.38±0.560.34±0.22c76.35

a P<0.05 vs. the vehicle control.

b P<0.01 vs. the vehicle control.

c P<0.001 vs. the vehicle control.

Discussion

A previous published study indicated that trifolirhizin had cytotoxicity in several cancer cells (19). Despite the cytotoxicity on human cells by previous reports, the markedly higher IC50 in our finding manifested the tissue specific effect of trifolirhizin on HEK293 (normal kidney cell) and L02 (normal liver cell). Therefore, trifolirhizin could be a potent safe anticancer candidate although the administration or the structure of trifolirhizin needs further efforts in future. In 2004, it was indicated that trifolirhizin induces apoptosis in HL-60 cells (23). However, the underlying apoptotic mechanism remains unclear. Thus, it was chosen as one candidate agent in our large scale natural anticancer compound screening for further efforts. The MKN45 cell is a typical gastric cancer cell commonly conducted as an anti-tumor model (24,25). Our assay indicated that trifolirhizin inhibited the proliferation of MKN45 gastric carcinoma cells in a time- and dose-dependent manner. Apoptosis hypothesis was further confirmed by flow cytometry data and visual contact under a microscope.

Many studies have indicated that EGFR-MAPK pathway has the ability to affect the Bcl-2 family (26). For instance, JNK can inactivate Bcl-2 while eliciting mitochondrial translocation of Bax (27). The Bcl-2 protein family is a crucial regulator of apoptosis (28). Bcl-2 protein family contain two main types of members: pro-apoptotic members Bax, anti-apoptotic members Bcl-2. The ratio of Bax/Bcl-2 leads to impaired apoptosis (29). In this study, imbalance in expression level of Bcl-2/Bax was clearly detected in the trifolirhizin treated MKN45 cells. MMP decreased significantly in trifolirhizin treated MKN45 cells. Loss of MMP contributes to the caspase pathway cascade (30). Caspases, a family of cysteine proteases, are important proteins in regulating the apoptotic response (31). Caspase-9 represents the intrinsic cascade pathway, while caspase-3 is the key executioner in apoptosis (32). Based on our western blot assay results, trifolirhizin induced the activation of caspase-9, caspase-3, and cleavage of PARP. Then apoptosis triggered cells to death. On the other hand, most cytotoxic agents usually inhibit cell proliferation via arresting the cycle in G1/S or G2/M phase (33,34). This arrest process is mediated with cyclin-dependent protein (Cdcs) with specific cyclin proteins. Trifolirhizin treatment leads to mediating accumulation in blocking of Cdc2 and inhibition of cyclin B resulting in a G2/M phase arrest.

We used the EGF (100 ng/ml) to re-activate EGFR signal pathway for the mechanism validation (35). The apoptosis ratio in trifolirhizin (30 µg/ml) combined with EGF using flow cytometry assay was decreased compared with trifolirhizin (30 µg/ml, Fig. 2). The cell cycle distributions were also transformed (30 µg/ml, Fig. 3). The EGFR protein levels were upregulated in the combination group (30 µg/ml, Fig. 5B). The examination manifested that EGFR signaling pathway may be paramount in the effect of trifolirhizin on MKN45 cells.

In conclusion, trifolirhizin induced apoptosis in MKN45 cancer cells in vitro and in vivo mediated via EGFR-MAPK pathways. Trifolirhizin also arrest G2/M cycle through impact on Cdc2/cyclin B complex (Fig. 4C). The results of this study implied that trifolirhizin may a feasible agent for anti-stomach tumor treatment, further scientific and therapeutic development should be established.

Acknowledgments

This study was supported by State Administration of Traditional Chinese Medicine 'Twelfth Five Year Plan' Key Specialty (Chinese Medicine Geriatrics). The Shanghai Key Laboratory of Clinical Geriatric Medicine provided important technical support.

References

1 

Tang X, Hu G, Xu C, Ouyang K, Fang W, Huang W, Zhang J, Li F, Wang K, Qin X, et al: HZ08 reverse the aneuploidy-induced cisplatin-resistance in gastric cancer by modulating the p53 pathway. Eur J Pharmacol. 720:84–97. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Saika K and Sobue T: Cancer statistics in the world. Gan To Kagaku Ryoho. 40:2475–2480. 2013.In Japanese. PubMed/NCBI

3 

Nomoto-Kojima N, Aoki S, Uchihashi K, Matsunobu A, Koike E, Ootani A, Yonemitsu N, Fujimoto K and Toda S: Interaction between adipose tissue stromal cells and gastric cancer cells in vitro. Cell Tissue Res. 344:287–298. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Fock KM: Review article: The epidemiology and prevention of gastric cancer. Aliment Pharmacol Ther. 40:250–260. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Hacker U and Lordick F: Current standards in the treatment of gastric cancer. Deutsche medizinische Wochenschrift. 140:1202–1205. 2015.In German.

6 

Safarzadeh E, Sandoghchian Shotorbani S and Baradaran B: Herbal medicine as inducers of apoptosis in cancer treatment. Adv Pharm Bull. 4(Suppl 1): 421–427. 2014.PubMed/NCBI

7 

Li HY, Zhang J, Sun LL, Li BH, Gao HL, Xie T, Zhang N and Ye ZM: Celastrol induces apoptosis and autophagy via the ROS/JNK signaling pathway in human osteosarcoma cells: An in vitro and in vivo study. Cell Death Dis. 6:e16042015. View Article : Google Scholar : PubMed/NCBI

8 

Zhang HH, Kuang S, Wang Y, Sun XX, Gu Y, Hu LH and Yu Q: Bigelovin inhibits STAT3 signaling by inactivating JAK2 and induces apoptosis in human cancer cells. Acta Pharmacol Sin. 36:507–516. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Xin Y, Yin F, Qi S, Shen L, Xu Y, Luo L, Lan L and Yin Z: Parthenolide reverses doxorubicin resistance in human lung carcinoma A549 cells by attenuating NF-κB activation and HSP70 up-regulation. Toxicol Lett. 221:73–82. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Lin LL, Hsia CR, Hsu CL, Huang HC and Juan HF: Integrating transcriptomics and proteomics to show that tanshinone IIA suppresses cell growth by blocking glucose metabolism in gastric cancer cells. BMC Genomics. 16:412015. View Article : Google Scholar : PubMed/NCBI

11 

Ye B, Li J, Li Z, Yang J, Niu T and Wang S: Anti-tumor activity and relative mechanism of ethanolic extract of Marsdenia tenacissima (Asclepiadaceae) against human hematologic neoplasm in vitro and in vivo. J Ethnopharmacol. 153:258–267. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Kocevski D, Du M, Kan J, Jing C, Lačanin I and Pavlović H: Antifungal effect of Allium tuberosum, Cinnamomum cassia, and Pogostemon cablin essential oils and their components against population of Aspergillus species. J Food Sci. 78:M731–M737. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Kim DW, Chi YS, Son KH, Chang HW, Kim JS, Kang SS and Kim HP: Effects of sophoraflavanone G, a prenylated flavonoid from Sophora flavescens, on cyclooxygenase-2 and in vivo inflammatory response. Arch Pharm Res. 25:329–335. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Hwang JS, Lee SA, Hong SS, Lee KS, Lee MK, Hwang BY and Ro JS: Monoamine oxidase inhibitory components from the roots of Sophora flavescens. Arch Pharm Res. 28:190–194. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Jung HJ, Kang SS, Hyun SK and Choi JS: In vitro free radical and ONOO- scavengers from Sophora flavescens. Arch Pharm Res. 28:534–540. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Fujise Y, Toda T and Itô S: Isolation of trifolirhizin from Ononis spinosa L. Chem Pharm Bull (Tokyo). 13:93–95. 1965. View Article : Google Scholar

17 

Zhang C, Ma Y, Gao HM, Liu XQ, Chen LM, Zhang QW, Wang ZM and Li AP: Non-alkaloid components from Sophora flavescens. Zhongguo Zhong Yao Za Zhi. 38:3520–3524. 2013.In Chinese.

18 

Abdel-Kader MS: Preliminary pharmacological study of the pterocarpans macckian and trifolirhizin isolated from the roots of Ononis vaginalis. Pak J Pharm Sci. 23:182–187. 2010.PubMed/NCBI

19 

Zhou H, Lutterodt H, Cheng Z and Yu LL: Anti-inflammatory and antiproliferative activities of trifolirhizin, a flavonoid from Sophora flavescens roots. J Agric Food Chem. 57:4580–4585. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Yang N, Liang B, Srivastava K, Zeng J, Zhan J, Brown L, Sampson H, Goldfarb J, Emala C and Li XM: The Sophora flavescens flavonoid compound trifolirhizin inhibits acetylcholine induced airway smooth muscle contraction. Phytochemistry. 95:259–267. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Hyun SK, Lee WH, Jeong DM, Kim Y and Choi JS: Inhibitory effects of kurarinol, kuraridinol, and trifolirhizin from Sophora flavescens on tyrosinase and melanin synthesis. Biol Pharm Bull. 31:154–158. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Yin T, Yang G, Ma Y, Xu B, Hu M, You M and Gao S: Developing an activity and absorption-based quality control platform for Chinese traditional medicine: Application to Zeng-Sheng-Ping (Antitumor B). J Ethnopharmacol. 172:195–201. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Aratanechemuge Y, Hibasami H, Katsuzaki H, Imai K and Komiya T: Induction of apoptosis by maackiain and trifolirhizin (maackiain glycoside) isolated from sanzukon (Sophora Subprostrate Chen et T. Chen) in human promyelotic leukemia HL-60 cells. Oncol Rep. 12:1183–1188. 2004.PubMed/NCBI

24 

Lindén SK, Driessen KM and McGuckin MA: Improved in vitro model systems for gastrointestinal infection by choice of cell line, pH, microaerobic conditions, and optimization of culture conditions. Helicobacter. 12:341–353. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Giard DJ, Aaronson SA, Todaro GJ, Arnstein P, Kersey JH, Dosik H and Parks WP: In vitro cultivation of human tumors: Establishment of cell lines derived from a series of solid tumors. J Natl Cancer Inst. 51:1417–1423. 1973.PubMed/NCBI

26 

Thomas S, Quinn BA, Das SK, Dash R, Emdad L, Dasgupta S, Wang XY, Dent P, Reed JC, Pellecchia M, et al: Targeting the Bcl-2 family for cancer therapy. Expert Opin Ther Targets. 17:61–75. 2013. View Article : Google Scholar

27 

Yu H, Zhang T, Cai L, Qu Y, Hu S, Dong G, Guan R, Xu X and Xing L: Chamaejasmine induces apoptosis in human lung adenocarcinoma A549 cells through a Ros-mediated mitochondrial pathway. Molecules. 16:8165–8180. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Llambi F and Green DR: Apoptosis and oncogenesis: Give and take in the BCL-2 family. Curr Opin Genet Dev. 21:12–20. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Bah N, Maillet L, Ryan J, Dubreil S, Gautier F, Letai A, Juin P and Barillé-Nion S: Bcl-xL controls a switch between cell death modes during mitotic arrest. Cell Death Dis. 5:e12912014. View Article : Google Scholar : PubMed/NCBI

30 

David R: Apoptosis: A lipid trigger of MOMP. Nat Rev Mol Cell Biol. 13:208–209. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Cohen GM: Caspases: The executioners of apoptosis. Biochem J. 326:1–16. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Brentnall M, Rodriguez-Menocal L, De Guevara RL, Cepero E and Boise LH: Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 14:322013. View Article : Google Scholar : PubMed/NCBI

33 

Carduner L, Picot CR, Leroy-Dudal J, Blay L, Kellouche S and Carreiras F: Cell cycle arrest or survival signaling through αv integrins, activation of PKC and ERK1/2 lead to anoikis resistance of ovarian cancer spheroids. Exp Cell Res. 320:329–342. 2014. View Article : Google Scholar

34 

Romanov V, Whyard TC, Waltzer WC, Grollman AP and Rosenquist T: Aristolochic acid-induced apoptosis and G2 cell cycle arrest depends on ROS generation and MAP kinases activation. Arch Toxicol. 89:47–56. 2015. View Article : Google Scholar

35 

Li L, Gao Y, Zhang L, Zeng J, He D and Sun Y: Silibinin inhibits cell growth and induces apoptosis by caspase activation, down-regulating survivin and blocking EGFR-ERK activation in renal cell carcinoma. Cancer Lett. 272:61–69. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 36 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lu X, Ma J, Qiu H, Yang L, Cao L and Shen J: Anti-proliferation effects of trifolirhizin on MKN45 cells and possible mechanism. Oncol Rep 36: 2785-2792, 2016
APA
Lu, X., Ma, J., Qiu, H., Yang, L., Cao, L., & Shen, J. (2016). Anti-proliferation effects of trifolirhizin on MKN45 cells and possible mechanism. Oncology Reports, 36, 2785-2792. https://doi.org/10.3892/or.2016.5125
MLA
Lu, X., Ma, J., Qiu, H., Yang, L., Cao, L., Shen, J."Anti-proliferation effects of trifolirhizin on MKN45 cells and possible mechanism". Oncology Reports 36.5 (2016): 2785-2792.
Chicago
Lu, X., Ma, J., Qiu, H., Yang, L., Cao, L., Shen, J."Anti-proliferation effects of trifolirhizin on MKN45 cells and possible mechanism". Oncology Reports 36, no. 5 (2016): 2785-2792. https://doi.org/10.3892/or.2016.5125