• 沒有找到結果。

Gypenosides Suppress Growth of Human Oral Cancer SAS Cells In Vitro and in a Murine Xenograft Model: The Role of Apoptosis Mediated by Caspase-Dependent and Caspase-Independent Pathways

N/A
N/A
Protected

Academic year: 2021

Share "Gypenosides Suppress Growth of Human Oral Cancer SAS Cells In Vitro and in a Murine Xenograft Model: The Role of Apoptosis Mediated by Caspase-Dependent and Caspase-Independent Pathways"

Copied!
7
0
0

加載中.... (立即查看全文)

全文

(1)

Gypenosides Suppress Growth of

Human Oral Cancer SAS Cells In

Vitro and in a Murine Xenograft

Model: The Role of Apoptosis Mediated by

Caspase-Dependent and Caspase-Independent Pathways

Kung-Wen Lu, MD, PHD

1

, Jung-Chou Chen, MD, PHD

1

, Tung-Yuan Lai, MD, PHD

1, 2

,

Jai-Sing Yang, PHD

2

, Shu-Wen Weng, MD

1

, Yi-Shih Ma, MD

1

, Hui-Yi Lin, PhD

1

,

Rick Sai-Chuan Wu, MD, PhD

2

, King-Chuen Wu, MD, PhD

2

, W. Gibson Wood, PhD

3

,

and Jing-Gung Chung, PhD

1,4

Abstract

Purpose. Gypenosides (Gyp) are the major components of Gynostemma pentaphyllum Makino. The authors

investigated the

effects of Gyp on cell morphology, viability, cell cycle distribution, and induction of apoptosis in human oral cancer SAS cells

and the determination of murine SAS xenograft model in vivo. Experimental design. Flow cytometry was used to quantify

the percentage of viable cells; cell cycle distribution; sub-G1 phase (apoptosis); caspase-3, -8, and -9 activity; reactive oxygen

species (ROS) production, intracellular Ca2+ determination; and the level of mitochondrial membrane potential

(ΔΨm).

Western blotting was used to examine levels of apoptosis-associated proteins, and confocal laser microscopy was used to

examine the translocation of proteins in cells. Results. Gyp induced morphological changes, decreased the percentage of

viable cells, caused G0/G1 phase arrest, and triggered apoptotic cell death in SAS cells. Cell cycle arrest induced by Gyp

was associated with apoptosis. The production of ROS, increased intracellular Ca2+ levels, and the depolarization

of ΔΨm

were observed. Gyp increased levels of the proapoptotic protein Bax but inhibited the levels of the antiapoptotic proteins

Bcl-2 and Bcl-xl. Gyp also stimulated the release of cytochrome c and Endo G. Translocation of GADD153 to the nucleus

was stimulated by Gyp. Gyp in vivo attenuated the size and volume of solid tumors in a murine xenograft model of oral

cancer. Conclusions. Gyp-induced cell death occurs through caspase-dependent and caspase-independent apoptotic signaling

pathways, and the compound reduced tumor size in a xenograft nu/nu mouse model of oral cancer.

Keywords

gypenosides, traditional Chinese medicine, caspase cascade, caspase independent, murine xenograft model, human oral

(2)

cancer SAS cells

Background and Introduction

Oral and pharyngeal cancers account for more than 300 000 cases annually worldwide, with men outnumbering women.1 In Taiwan, a 2009 report from the Department of Health, ROC Taiwan, indicated that 9.7 individuals per 100 000 die annually from oral cancer. Treatments for oral cancer, including surgery, radiotherapy, and current chemotherapeutic options, are inadequate, and there is a great need to

identify new agents and novel targets for treating oral cancer.2 To that end, complementary and alternative medicine (CAM) is gaining attention in treating various cancers.3,4 Several studies have reported that compounds from traditional medicines and herbs are being widely investigated as potential therapeutic agents.5-7 In the United States, approximately 36% of the population use alternative medicines based 1China Medical University, Taichung, Taiwan

2China Medical University Hospital, Taichung, Taiwan

3University of Minnesota, VA Medical Center, Minneapolis, MN, USA

4Asia University, Wufeng, Taichung, Taiwan

Corresponding Author:

Jing-Gung Chung, Department of Biological Science and

Technology, China Medical University, No 91, Hsueh-Shih Road, Taichung 40402, Taiwan

Email: jgchung@mail.cmu.edu.tw

on a report from the National Institute of Health’s National Center for Alternative and Complementary Medicine. Cancer patients use CAM extensively as a mode of treatment or as a means to reduce the side effects of conventional treatments.8-10

Among the Chinese population, Gynostemma pentaphyllum Makino has been a popular folk medicine for centuries, and gypenosides (Gyp) in this plant species have been

used to treat hepatitis,11 hyperlipoproteinemia,12,13 cardiovascular disease,14 and cancer.15 Gyp has biological activities

that include anti-inflammatory,16 antithrombotic,17 antioxidative, 18 and anticancer1,19,20 actions. We have reported that

Gyp induced apoptosis in human lung cancer A549 cells,21 colon cancer colo 205 cells,22 and tongue cancer SCC-4 cells23 and also inhibited migration and invasion of human tongue cancer SCC-4 cells.24 Other investigators found that Gyp induced apoptosis in human hepatoma cells.1 However,

there have not been any reports on Gyp-induced apoptosis in human oral cancer SAS cells in vitro or in vivo. Therefore, we investigated the effects of Gyp on induction of apoptosis in vitro and the determination of the SAS murine xenograft model in vivo.

Materials and Methods

Chemicals, Reagents, and Cell Culture

Gyp was kindly provided by Dr Jung-Chou Chen (Department of Chinese Medicine, China Medical University, Taichung, Taiwan).25 Dimethyl sulfoxide (DMSO), propidium iodide (PI), potassium phosphates, ribonuclease-A, Triton X-100, Tris–HCl, and trypan blue were obtained from Sigma

Chemical Co (St Louis, MO). 2,7-Dichlorodihydrofluorescein diacetate, DiOC6, and Fluo-3/AM were obtained from Molecular Probes/Invitrogen Corp (Eugene, OR). Dulbecco’s modified Eagle’s medium (DMEM), L-glutamine, fetal bovine serum (FBS), penicillin-streptomycin, and trypsin-EDTA were obtained from GIBCO BRL/Invitrogen Corp (Grand Island, NY). The SAS cell line (human oral squamous cell carcinoma) was obtained from Dr Pei-Jung Lu (Graduate Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan). Cells were cultured in DMEM containing 10% FBS, 2 mM L-glutamine, 100 U/mL penicillin, and 100 μg/mL streptomycin in 75 cm2 tissue culture

flasks at 37°C under a humidified 5% CO2and 95% air atmosphere as we have previously reported.22

In Vitro Studies

Assessment of cell morphology and viability. Gyp was prepared

and dissolved in DMSO. Cells (2 × 105 cells/well)

were plated in 12-well plates in 2 mL DMEM and incubated at 37°C for 24 hours. Cells were then treated with 0, 60, 90, 120, 150, and 180 μg/mL Gyp for 24 hours. DMSO was used as a vehicle control. At the end of the incubation period, cells were photographed with a phase-contrast microscope. They were then harvested, stained with PI (5 μg/mL) and analyzed by flow cytometry (Becton-Dickinson, San Jose, CA) as previously described.22,26,27

Flow cytometry analysis of sub-G1 phase in SAS cells. Cells

were incubated with 0, 60, 90, 120, 150, and 180 μg/mL Gyp for different time periods, after which time they were harvested by centrifugation and stained with PI (5 μg/mL). The percentage of cells in the sub-G1 (apoptosis), G0/G1, S, and G2/M phases were determined by flow cytometry as previously described.22,28

(3)

4,6-Diamidino-2-phenylindole dihydrochloride (DAPI) staining for apoptotic cell death. SAS cells were treated with or without

Gyp (0, 60, 90, 120, 150, and 180 μg/mL) for 48 hours. They were then isolated, stained with DAPI, and photographed using a fluorescence microscope, as previously

described.28,29

Detections of reactive oxygen species (ROS), intracellular Ca2+ levels, and mitochondrial membrane potential (ΔΨ m) in

SAS cells. SAS cells were treated with 180 μg/mL Gyp for

0.5, 1, 3, 6, 12, 24, and 48 hours. The cells were harvested and washed twice, resuspended in 500 μL of

,7-dichlorodihydrofluorescein diacetate (10 μM) for determining changes of ROS, in 500 μL of Fluo-3/AM (2.5 μg/mL) for staining of intracellular Ca2+, and in 500 μL of DiOC6 (1 μmol/L) for determining ΔΨm. The cells were incubated at 37°C for 30 minutes and were analyzed by flow cytometry.28,30,31

Confocal laser scanning microscopy for protein translocation in SAS cells. SAS cells in DMEM were plated on 4-well

chamber slides and treated without or with 180 μg/mL Gyp for 24 hours. Cells were then fixed in 4% formaldehyde in

PBS (phosphate-buffered saline) for 15 minutes and permeabilized with 0.3% Triton-X 100 in PBS for 1 hour, with

nonspecific binding sites being blocked using 2% BSA. Fixed cells were then incubated with primary antibodies to AIF, Endo G, cytochrome c, and GADD153 (1:100 dilution; green fluorescence) overnight and were then stained by a secondary antibody (FITC-conjugated goat antimouse IgG at 1:100 dilution), followed by DNA and mitochondria staining with PI and rhodamine 123 (red fluorescence). Photomicrographs were obtained using a Leica TCS SP2 Confocal Spectral Microscope.22,28

Determination of levels of certain proteins associated with apoptotic cell death. Cells were incubated with 180 μg/mL

Gyp for 0, 6, 12, 24, 48, and 72 hours, isolated, and lysed, and specific protein levels associated with cell cycle arrest and apoptosis were determined. All samples were separated by sodium dodecyl sulfate polyacrylamide (SDS-PAGE) gel electrophoresis and Western blotting as previously described.22,28,29

Real-time polymerase chain reaction (PCR) for caspase-3, -8, -9 mRNA levels. Cells were incubated with 180 μg/mL Gyp

for 0 and 48 hours. Total RNA was extracted using the Qiagen RNeasy Mini Kit (Qiagen, Inc, Valencia, CA), and

RNA samples were then reverse transcribed with the High

Capacity cDNA Reverse Transcription Kit according to the standard protocol of the supplier (Applied Biosystems, Carlsbad, CA) as previously described.32 Quantitative PCR was performed on 200 nM of forward (F) and reverse (R) primers (caspase-3, F-CAGTGGAGGCCGACTTCTTG, R TGGCACAAAGCGACTGGAT; caspase-8, F-GGATG GCCACTGTGAATAACTG, R-TCGAGGACATCGCTCT CTCA; caspase-9, F-TGTCCTACTCTACTTTCCCAGGT TTT, R-GTGAGCCCACTGCTCAAAGAT; GAPDH, F-AC ACCCACTCCTCCACCTTT, R-TAGCCAAATTCGTTGTC ATACC). Each assay was run on an Applied Biosystems 7300 Real-Time PCR system in triplicate, and fold changes in

expression were derived using the comparative CT method.27,33

Determinations of caspase-3, -8, and -9 activity. Cells were

treated with 180 μg/mL of Gyp for 0, 6, 12, 24, 48, and 72 hours. They were then collected by centrifugation; 50 μL of each 10 μM substrate solution (PhiPhiLux-G1D2 for caspase-3, CaspaLux8-L1D2 for caspase-8, and CaspaLux9-M1D2 for caspase-9; OncoImmunin, Inc, Gaithersburg, MD)

was added, and individual caspase activity was analyzed according to the manufacturer’s instructions using flow cytometry as previously described.34,35 Cells were also treated with 180 μg/mL Gyp in the presence or absence of 10 μM of caspase-9 inhibitor (Z-IETD-FMK), 10 μM of caspase-8 inhibitor (Z-IETD-FMK), 10 μM of caspase-3 inhibitor (Z-DEVD-FMK), and 10 μM of general caspase inhibitor (Z-VAD-FMK; R&D Systems, Minneapolis, MN), or 5 mM of NAC (Sigma Chemical Co) for ROS and 5 μM of BAPTA (Ca2+ chelator, Molecular Probes/Invitrogen Corp.) for intracellular Ca2+. Cell viability was determined as previously described.21,36

In Vivo Studies

SAS mouse xenograft model. A total of 18 male athymic

BALB/cnu/nu nude mice 4 to 6 weeks of age were purchased from the National Laboratory Animal Center of National Applied Research Laboratories (Taipei, Taiwan). Mice were maintained on a commercial diet and water ad libitum. For the injections, SAS cells were resuspended in serum-free DMEM. The cell suspension (5 × 106 cells/0.2 mL) was subcutaneously injected into the flanks of mice for an 8-day incubation period. After xenografts reached volumes of 200 mm3, treatment was initiated. Body weight and tumor size were measured every 2 days. Mice bearing tumors were randomly divided into 3 treatment groups and

(4)

injected every 2 days in the morning with 30 μL of

DMSO control vehicle, 2 mg/kg doxorubicin, and 20 mg/kg Gyp. The measurement of tumor volume was based on the following formula: Tumor volume (mm3) = L × W2/2 (L is the length and W the width). At the end of the 28 days of

treatment, mice were killed, and tumors were removed, measured, and weighted as previously described.33,37,38 All animal studies were conducted according to institutional guidelines (Affidavit of Approval of Animal Use Protocol) approved by the Institutional Animal Care and Use Committee (IACUC) of China Medical University (Taichung, Taiwan).

Statistical Analysis

Data of control and experimental groups were expressed as mean ± standard deviation for at least 3 separate experiments. Statistical analyses of the data were performed using

Student’s t test and 1-way analysis of variance (ANOVA). Statistical significance was set at P < .05.

Results

In Vitro Studies

Effects of Gyp on morphology, viability, cell cycle distribution, and sub-G1 phase of SAS cells. Cells were morphologicallyaltered

by Gyp treatment as shown in Figure 1A. There were fewer viable cells as incubation time and Gyp concentration increased (Figure 1B). It can be seen in Figure 1C that there was an increase in the percentage of cells in G0/G1 (enhanced G0/G1 peak) and a decrease in the percentage of cells in the S phase. The sub-G1 group also appeared in the cell cycle (Figure 1C), and increasing concentrations of Gyp led to an increase in G0/G1- and sub-G1 phases.

Gyp-induced apoptosis in SAS cells. Induction of apoptosis

by Gyp in SAS cells was confirmed by DAPI staining, as seen in Figure 2A, which showed that Gyp induced nuclei condensation. These effects were time dependent as noted in Figure 2B. Higher concentrations of Gyp resulted in a greater number of apoptotic cells being stained.

Gyp induced production of ROS and intracellular Ca2+ and reduction of mitochondrial membrane potential (ΔΨm) in SAS cells. Gyp induced ROS production quite early, and this was

time dependent, as seen in Figure 3A. After 0.5 hours of incubation, there was a decrease in ROS levels in the Gyp group. Gyp stimulated intracellular Ca2+ release, which was time dependent (Figure 3B), and reduced the levels of ΔΨm in a time-dependent manner (Figure 3C). Both ROS and intracellular Ca2+ were increased by Gyp (Figure 3). We next determined if the ROS scavenger (NAC) and Ca2+ chelator

(BAPTA) could inhibit effects of Gyp on cell viability and apoptosis. It can be seen in Figure 3 (panels A, B, C, and D) that Gyp increased the levels of ROS, but NAC protected Gyp-treated SAS cells against induction of cell death

(Figure 4A) and reduction of apoptotic cells (Figure 4C). Gyp increased intracellular Ca2+, but BAPTA was protective (Figure 4B) and reduced apoptotic SAS cell numbers

(Figure 4D). NAC and BAPTA significantly blocked Gypinduced cell death.

Gyp increased the translocation of proteins in SAS cells. The

results from confocal laser microscopy indicated that the

protein levels of AIF (Figure 5A), Endo G (Figure 5B), cytochrome c (Figure 5C), and GADD153 (Figure 5D) were

increased when compared with the levels in controls. It can be seen that cytochrome c is released from mitochondria, as seen by the higher green fluorescence intensity. AIF, Endo G, and GADD153 trafficked to nuclei, which indicated higher fluorescence in SAS cells.

Effects of Gyp on levels of proteins associated with cell cycle and apoptosis. Results are presented in Figures 6A, 6B, 6C,

6C, and 6D (A: chk2, p53, p21, Cdc25A, Cyclin E, Cyclin A, Cdk2; B: Bax, Bcl-2, puma, cytochrome c, Endo G, AIF, XIAP, CAD, Caspase-9, Caspase-3, caspase-7; C: TRAIL, Bid, Fas, FasL; D: ATF-4, GRP78, and GADD153). Gyp treatment reduced the levels of cdc25A, cyclin E and A and cdk2 (Figure 6A), Bcl-2 and XIAP (Figure 6B), and Bid (Figure 6C), whereas the levels of p53 and p21 (Figure 6A)

were increased. Gyp increased protein levels of Bax, puma, cytochrome c, Endo G, AIF, CAD, caspase-9 and caspase-3 (Figure 6B), TRAIL, Fas and FasL (Figure 6C), ATF-4, GRP78, and GADD153 (Figure 6D) but reduced levels of Bcl-2 and XIAP (Figure 6B) and Bid (Figure 6C). Expression levels of mRNA of caspase-3, -8, and -9 were increased after Gyp treatment (Figure 6E).

Gyp stimulated activity of caspase-3, -8, and -9. Data seen in

Figure 7 indicate that Gyp increased the activity of caspase-3, caspase-8, and caspase-9, and these effects were time dependent. To determine whether caspase activation was involved

in apoptotic cell death, cells were pretreated with inhibitors of caspase-3, caspase-8, and caspase-9 and a general inhibitor of caspases. As shown in Figures 8A, 8B, 8C, and 8D, these inhibitors significantly blocked Gyp-triggered cell death. Gyp

inhibited tumor size in a xenograft mouse model. Results

from our in vitro studies indicated that Gyp induced cell cycle arrest and apoptosis in SAS cells through mitochondrialdependent

(5)

and -independent pathways. We next examined

the in vivo antitumor activities of Gyp in BALB/cnu/nu nude mice injected with SAS cells and treated with Gyp and doxorubicin (positive control). Representative tumors in the xenograft mice treated with or without Gyp are shown in Figure 9A. Treatment with Gyp and doxorubicin did not significantly alter body weight (Figures 9B and 9D). Gyp significantly (P < .01) decreased the tumor weight by 34%, compared with controls, after treatment from the 10th to the 28th day, as shown in Figure 9C. In Figures 9A and 9C, it can be seen that doxorubicin (2 mg/kg) also significantly reduced tumor mass by ~49% after 28 days of treatment. Overall, the tumors in treatment groups were significantly smaller than those in the control group.

Discussion

We had previously reported that Gyp induced apoptosis in human colon cancer cells, which was mitochondria dependent and involved caspase-3 activation,23 and in human

tongue cancer SCC-4 cells through endoplasmic reticulum (ER) stress and mitochondria-dependent pathways.22 Effects of Gyp on human oral cancer cells have not been examined, and the purpose of the present study was to determine whether or not Gyp induced S phase arrest and apoptosis in human oral cancer SAS cells. We found that Gyp induced cell death through cell cycle arrest and induction of apoptosis. Apoptosis followed Gyp-induced S phase arrest. Gyp may have therapeutic efficacy in the treatment of oral cancers.

Gyp-induced morphological changes and reduced viability

of SAS cells were dose and time dependent. Gyp induced cell cycle arrest, and the compound altered levels of proteins

associated with the S phase in SAS cells. Protein levels of CDK2, Cyclin A, and Cyclin E were reduced. Furthermore, in model cells, it has been shown that upregulation of p53 was sufficient to activate protein kinase c–mediated p53 gene transcription,

induce G1 phase arrest, and promote cellular repair

mechanisms.39,40 Gyp-induced apoptosis in SAS cells was also confirmed by DAPI staining and DNA gel electrophoresis (data not shown). Gyp-induced apoptosis in SAS cells and these effects were dose and time dependent. It is well documented that the Bcl-2 family plays an important role in apoptosis

with respect to both antiapoptotic11 and proapoptotic41 signaling. The ratio of Bcl-2 to Bax is an indicator of sensitivity or resistance to apoptotic stimuli.42

Figure 3A indicates that Gyp promoted ROS production in the earlier time periods of treatment of SAS cells. Much evidence suggests that ROS play an important role in apoptosis induced by certain chemopreventive agents via the

engagement of downstream proteins involved in the execution of apoptosis.42-44 The intracellular generation of H2O2 (the most stable ROS) has been shown to be an important

mediator of apoptosis, including exogenous addition of H2O2.45 H2O2 can diffuse into the mitochondria and may cause damage to the mitochondrial membrane.33,45 Results from Figure 3B indicate that Gyp increased Ca2+ levels at earlier time points, suggesting that Gyp treatment induced

ER stress, which elicited a rise in intracellular Ca2+ and subsequent mitochondrial membrane depolarization (decreased the levels of ΔΨ m) in SAS cells. This finding is in agreement with other reports that Ca2+-mediated signaling is involved in Gyp-induced

apoptosis.23,24

In the present study, Gyp increased Bax levels but reduced Bcl-2 levels. Gyp decreased the levels of ΔΨm and promoted

the release of cytochrome c, AIF, and Endo G and increased both the levels and activity of caspase-9 and -3 resulting in apoptosis. Gyp-induced cell survival or death may occur through the interaction of Bcl-2- and p53-mediated mechanisms followed by caspase-9 and -3 activation. One conclusion is that Gyp-induced apoptosis involves multisignaling

pathways that are caspase dependent and independent. To further understand the role of Gyp we used an in vivo SAS xenograft mouse model and found that Gyp decreased the weight and size of SAS cells tumors, which is certainly in agreement with our in vitro results. Findings of the present study provide new insight into possible multiple signaling pathways of Gyp-induced apoptosis in human oral cancer SAS cells. It is well known that certain products from plants are known to

induce apoptosis in tumor cells.43-45 Understanding the mech-anisms

of Gyp on tumor cells may provide valuable information that can be used in cancer therapy and prevention.

Declaration of Conflicting Interests

The author(s) declared no potential conflicts of interest with respect to the authorship and/or publication of this article.

Funding

The author(s) disclosed receipt of the following financial support for the research and/or authorship of this article: this work was supported by Grant DOH99-TD-C-111-005 from Department of Health, Executive Yuan, ROC (Taiwan).

(6)

1. Wang QF, Chen JC, Hsieh SJ, Cheng CC, Hsu SL. Regulation of Bcl-2 family molecules and activation of caspase cascade involved in gypenosides-induced apoptosis in human hepatoma cells. Cancer Lett. 2002;183:169-178.

2. Ichimiya Y, Fuwa N, Kamata M, et al. Treatment results of stage I oral tongue cancer with definitive radiotherapy. Oral

Oncol. 2005;41:520-525.

3. Brauer JA, El Sehamy A, Metz JM, Mao JJ. Complementary and alternative medicine and supportive care at leading cancer centers: a systematic analysis of websites. J Altern Complement

Med. 2010;16:183-186.

4. Lewith G, Britten N, Paterson C. Complementary and alternative medicine (CAM) and cancer: the kind face of complementary medicine. Int J Surg. 2010;8:81.

5. Powell CB, Fung P, Jackson J, et al. Aqueous extract of herba Scutellaria barbatae, a chinese herb used for ovarian cancer, induces apoptosis of ovarian cancer cell lines. Gynecol Oncol. 2003;91:332-340.

6. Rozanova N, Zhang JZ, Heck DE. Catalytic therapy of cancer with ascorbate and extracts of medicinal herbs [published

online ahead of print December 26, 2007]. Evid Based Complement

Alternat Med. 2010;7: 203–212.

7. Kaur M, Mandair R, Agarwal R, Agarwal C. Grape seed extract induces cell cycle arrest and apoptosis in human colon carcinoma cells. Nutr Cancer. 2008;60(suppl 1):2-11.

8. Richardson MA, Sanders T, Palmer JL, Greisinger A, Singletary SE. Complementary/alternative medicine use in a comprehensive cancer center and the implications for oncology.

J Clin Oncol. 2000;18:2505-2514.

9. Dy GK, Bekele L, Hanson LJ, et al. Complementary and alternative medicine use by patients enrolled onto phase I clinical

trials. J Clin Oncol. 2004;22:4810-4815.

10. Hlubocky FJ, Ratain MJ, Wen M, Daugherty CK. Complementary and alternative medicine among advanced cancer

patients enrolled on phase I trials: a study of prognosis, quality of life, and preferences for decision making. J Clin Oncol. 2007;25:548-554.

11. Lin CC, Huang PC, Lin JM. Antioxidant and hepatoprotective effects of Anoectochilus formosanus and Gynostemma pentaphyllum.

Am J Chin Med. 2000;28:87-96.

12. la Cour B, Molgaard P, Yi Z. Traditional Chinese medicine in treatment of hyperlipidaemia. J Ethnopharmacol. 1995; 46:125-129.

13. Yu R, Wang DS, Zhou H. Clinical and experimental study on effects of yinchen wuling powder in preventing and treating

hyperlipoproteinemia. Zhongguo Zhong Xi Yi Jie He Za Zhi. 1996;16:470-473.

14. Purmova J, Opletal L. Phytotherapeutic aspects of diseases of the cardiovascular system. 5. Saponins and possibilities of their use in prevention and therapy. Ceska Slov Farm. 1995;44:246-251.

15. Hou J, Liu S, Ma Z, Lang X, Wang J, Liang Z. Effects of Gynostemma

pentaphyllum makino on the immunological function

of cancer patients. J Tradit Chin Med. 1991;11:47-52.

16. Li L, Jiao L, Lau BH. Protective effect of gypenosides against oxidative stress in phagocytes, vascular endothelial cells and liver microsomes. Cancer Biother. 1993;8:263-272.

17. Tan H, Liu ZL, Liu MJ. Antithrombotic effect of Gynostemma

pentaphyllum. Zhongguo Zhong Xi Yi Jie He Za Zhi.

1993;13:278-280, 261.

18. Hu L, Chen Z, Xie Y. New triterpenoid saponins from Gynostemma pentaphyllum. J Nat Prod. 1996;59:1143-1145.

19. Wang C, Wang X, Li Y, Deng S, Jiang Y, Yue L. A preliminary observation of preventive and blocking effect of Gynostemma

pentaphyllum (Thunb) Makino on esophageal cancer in rats. Hua Xi Yi Ke Da Xue Xue Bao. 1995;26:430-432.

20. Zhou Z, Wang Y, Zhou Y. The effect of Gynostemma pentaphyllum mak (GP) on carcinogenesis of the golden hamster

cheek pouch induced by DMBA. Zhonghua Kou Qiang Yi Xue

Za Zhi. 1996;31:267-270.

21. Lu HF, Chen YS, Yang JS, et al. Gypenosides induced G0/G1 arrest via inhibition of cyclin E and induction of apoptosis via activation of caspases-3 and -9 in human lung cancer A-549 cells. In Vivo. 2008;22:215-221.

22. Chen JC, Lu KW, Tsai ML, et al. Gypenosides induced G0/G1 arrest via CHk2 and apoptosis through endoplasmic reticulum stress and mitochondria-dependent pathways in human tongue cancer SCC-4 cells. Oral Oncol. 2009;45:273-283. 23. Chen JC, Lu KW, Lee JH, Yeh CC, Chung JG. Gypenosides induced apoptosis in human colon cancer cells through the mitochondria-dependent pathways and activation of caspase-3.

Anticancer Res. 2006;26:4313-4326.

24. Lu KW, Tsai ML, Chen JC, et al. Gypenosides inhibited invasion and migration of human tongue cancer SCC4 cells

through down-regulation of NFkappaB and matrix metalloproteinase-9. Anticancer Res. 2008;28:1093-109metalloproteinase-9.

25. Hsu HY, Yang JS, Lu KW, et al. An experimental study on the antileukemia effects of gypenosides in vitro and in vivo [published online ahead of print August 11, 2010]. Integr Cancer

(7)

26. Lin CC, Yang JS, Chen JT, et al. Berberine induces apoptosis in human HSC-3 oral cancer cells via simultaneous activation of the death receptor-mediated and mitochondrial pathway.

Anticancer Res. 2007;27:3371-3378.

27. Ho YT, Yang JS, Li TC, et al. Berberine suppresses in vitro migration and invasion of human SCC-4 tongue squamous cancer cells through the inhibitions of FAK, IKK, NF-kappaB, u-PA and MMP-2 and -9. Cancer Lett. 2009;279:155-162. 28. Lin CC, Lin SY, Chung JG, Lin JP, Chen GW, Kao ST. Downregulation

of cyclin B1 and up-regulation of Wee1 by berberine promotes entry of leukemia cells into the G2/M-phase of the cell cycle. Anticancer Res. 2006;26:1097-1104.

29. Li TM, Chen GW, Su CC, et al. Ellagic acid induced p53/p21 expression, G1 arrest and apoptosis in human bladder cancer T24 cells. Anticancer Res. 2005;25:971-979.

30. Lu CC, Yang JS, Huang AC, et al. Chrysophanol induces necrosis through the production of ROS and alteration of ATP levels in J5 human liver cancer cells. Mol Nutr Food Res. 2010;54:967-976.

31. Lu HF, Wang HL, Chuang YY, et al. Danthron induced apoptosis through mitochondria- and caspase-3-dependent pathways

in human brain glioblastoma multiforms GBM 8401 cells.

Neurochem Res. 2010;35:390-398.

32. Chung JG, Yeh KT, Wu SL, et al. Novel transmembrane GTPase of non-small cell lung cancer identified by mRNA differential display. Cancer Res. 2001;61:8873-8879.

33. Ji BC, Hsu WH, Yang JS, et al. Gallic acid induces apoptosis via caspase-3 and mitochondrion-dependent pathways in vitro and suppresses lung xenograft tumor growth in vivo. J Agric

Food Chem. 2009;57:7596-7604.

34. Yang JS, Chen GW, Hsia TC, et al. Diallyl disulfide induces apoptosis in human colon cancer cell line (COLO 205) through the induction of reactive oxygen species, endoplasmic reticulum stress, caspases casade and mitochondrial-dependent pathways.

Food Chem Toxicol. 2009;47:171-179.

35. Chiu TH, Lai WW, Hsia TC, et al. Aloe-emodin induces cell death through S-phase arrest and caspase-dependent pathways in human tongue squamous cancer SCC-4 cells. Anticancer

Res. 2009;29:4503-4511.

36. Kuo HM, Tsai HC, Lin YL, et al. Mitochondrial-dependent caspase activation pathway is involved in baicalein-induced apoptosis

in human hepatoma J5 cells. Int J Oncol. 2009;35:717-724. 37. Yang SF, Yang WE, Chang HR, Chu SC, Hsieh YS. Luteolin induces apoptosis in oral squamous cancer cells. J Dent Res.

2008;87:401-406.

38. Ho YT, Yang JS, Lu CC, et al. Berberine inhibits human tongue squamous carcinoma cancer tumor growth in a murine xenograft model. Phytomedicine. 2009;16:887-890.

39. Wahl GM, Carr AM. The evolution of diverse biological responses to DNA damage: insights from yeast and p53. Nat

Cell Biol. 2001;3:E277-E286.

40. Bullock AN, Fersht AR. Rescuing the function of mutant p53.

Nat Rev Cancer. 2001;1:68-76.

41. Oltvai ZN, Milliman CL, Korsmeyer SJ. Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates

programmed cell death. Cell. 1993;74:609-619.

42. Oltvai ZN, Korsmeyer SJ. Checkpoints of dueling dimers foil death wishes. Cell. 1994;79:189-192.

43. Androutsopoulos V, Arroo RR, Hall JF, Surichan S, Potter GA. Antiproliferative and cytostatic effects of the natural product eupatorin on MDA-MB-468 human breast cancer cells due to CYP1-mediated metabolism. Breast Cancer Res. 2008; 10:R39.

44. Jo H, Loison F, Hattori H, Silberstein LE, Yu H, Luo HR. Natural product Celastrol destabilizes tubulin heterodimer and facilitates mitotic cell death triggered by microtubuletargeting anti-cancer drugs. PLoS One. 2010;5:e10318.

45. Wang Z, Desmoulin S, Banerjee S, et al. Synergistic effects of multiple natural products in pancreatic cancer cells. Life Sci. 2008;83:293-300.

參考文獻

相關文件

Incubation with bisphosphonates reduced the cell count, but only became significant for zoledronate at 50 µmol (p=0.028) compared to the control; incubation with 50 µmol

原文題目(出處): Bisphosphonates: restrictions for vasculogenesis and angiogenesis: inhibition of cell function of endothelial progenitor cells and mature endothelial cells

(E) Varying numbers of tumour cells expressing cancer antigen 125 are present in the lining of tubules (objective magnification ¥10).. The arrowed cells show strong nuclear

42 Thus, the prevention of oral cancer and its associated morbidity and mortality, hinges upon the early detection of oral precancerous lesions, allowing for histological evaluation

The aggressiveness of the lesion and destructive growth noted in the present case, along with fascicular pattern of spindle cells in a mixed inflammatory infiltrate ruled out

potential agent to prevent and treat oral cancer... Structure and pharmacokinetics of

 Diagnosis is confirmed by histopathology showing the presence of Antoni A(cellular fascicular) and Antoni B (myxoid; vacuolated) cells, nuclear palisading, whirling of cells

Background: Gold standard for the diagnosis of oral dysplasia (OD) oral squamous cell carcinoma (OSCC) and malignant lesions is the histological examination.. Several