Pien Tze Huang inhibits the proliferation of human colon carcinoma cells by arresting G1/S cell cycle progression

  • Authors:
    • Aling Shen
    • Fei Hong
    • Liya Liu
    • Jiumao Lin
    • Lihui  Wei
    • Qiaoyan  Cai
    • Zhenfeng Hong
    • Jun Peng
  • View Affiliations

  • Published online on: July 19, 2012     https://doi.org/10.3892/ol.2012.811
  • Pages: 767-770
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pien Tze Huang (PZH), a well-known traditional Chinese formula prescribed 450 years ago in the Ming Dynasty, has been used in China and Southeast Asia for centuries as a folk remedy for various types of cancer, including colorectal cancer (CRC). Recently, we reported that PZH is capable of inhibiting colon cancer growth both in vivo and in vitro via the promotion of apoptosis and inhibition of tumor angiogenesis. To elucidate the mechanism of the tumoricidal activity of PZH, its effect on the proliferation of human colon carcinoma Caco-2 cells was evaluated and the underlying molecular mechanism was investigated. Results showed that PZH inhibited Caco-2 cell viability and survival in a dose‑ and/or time-dependent manner. In addition, PZH treatment was found to block the G1/S cell cycle progression. Moreover, PZH suppressed the mRNA and protein expression of pro‑proliferative Cyclin D1 and CDK4. Findings of the present study suggest that inhibition of cell proliferation via the G1/S cell cycle arrest is a potential mechanism by which PZH can be effective in the treatment of cancer.

Introduction

Colorectal cancer (CRC) is the second most common cause of cancer-related mortality in Western societies (1). Despite recent advances in CRC treatment, 5-fluorouracil (5-FU)-based regimens continue to be the international standard chemotherapy for patients with advanced CRC (2). However, drug resistance profoundly limits the effectiveness of current CRC cancer chemotherapies (3). Moreover, application of 5-FU-based regimens is often coupled with serious toxicity and side-effects such as anemia, leucopenia, thrombocytopenia and peripheral neuropathy (4,5). Therefore, it is essential to develop safer agents for the chemotherapeutic treatment of CRC. Traditional Chinese medicine (TCM), which has relatively few side-effects, plays an important role in primary health care in China and has recently been recognized by Western countries as a key source for revealing novel lead molecules for modern drug discovery. Clinical practice has also shown that many traditional Chinese medicines possess antitumor activities, which provide insight into new therapeutic strategies for cancer treatment (614).

Cancer cells are characterized by an uncontrolled increase in cell proliferation (15). Eukaryotic cell proliferation is primarily regulated by the cell cycle. G1/S transition is one of the two main checkpoints of the cell cycle (16), which is responsible for initiation and completion of DNA replication. G1/S progression is strongly regulated by Cyclin D1, which exerts its function by forming an active complex with its major catalytic partners, such as CDK4 (17). An unchecked or hyperactivated Cyclin D1/CDK4 complex often leads to uncontrolled cell division and malignancy (1821). Therefore, inhibiting excessive proliferation of tumor cells by blocking Cyclin D1/CDK4-mediated G1/S progression is one of the key approaches for the development of anticancer drugs.

Pien Tze Huang (PZH) is a well-known traditional Chinese formula that was first prescribed 450 years ago in the Ming Dynasty, with properties of heat-clearing and detoxification (22). In the TCM system, accumulation of toxic dampness and heat is a major pathogenic factor of cancer, therefore clearing heat and detoxification is a principle of anticancer treatment. For this reason, PZH has been used in China and Southeast Asia for centuries as a folk remedy for various types of cancer. Modern pharmacological studies have proposed that PZH exhibits therapeutic effects in clinical trials of tumors, such as hepatocellular carcinoma and colon cancer (23,24). In addition, in experimental animals PZH inhibits the growth of Ehrlich-Ascites tumor, gastric carcinoma, and hepatoma (25). Moreover, we recently reported that PZH is able to inhibit colon cancer growth both in vivo and in vitro via the promotion of apoptosis and inhibition of tumor angiogenesis (2628). To elucidate the mechanism of the tumoricidal activity of PZH, we evaluated its effect on the proliferation of the human colon carcinoma cell line Caco-2 and investigated the underlying molecular mechanism.

Materials and methods

Materials and reagents

RPMI-1640, fetal bovine serum (FBS), penicillin-streptomycin, Trypsin-EDTA, and TRIzol reagent were purchased from Invitrogen (Carlsbad, CA, USA). SuperScript II reverse transcriptase was obtained from Promega (Madison, WI, USA). Cyclin D1, CDK4 and β-actin antibodies, and horseradish peroxidase (HRP)-conjugated secondary antibodies were purchased from Cell Signaling (Beverly, MA, USA). Any other chemicals, unless otherwise stated, were obtained from Sigma-Aldrich (St. Louis, MO, USA).

Preparation of PZH

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

Cell culture

Human colon carcinoma Caco-2 cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). Caco-2 cells were grown in RPMI-1640 containing 10% (v/v) FBS, and 100 U/ml penicillin and 100 μg/ ml streptomycin. Cells were cultured at 37°C, in a 5% CO2 humidified environment.

Assessment of cell viability

Cell viability was assessed by MTT colorimetric assay. Caco-2 cells were seeded into 96-well plates at a density of 5×103 cells/well in 0.1 ml medium. The cells were treated with various concentrations of PZH for different periods of time. At the end of the treatment, 100 μl MTT (0.5 mg/ml in PBS) were added to each well, and the samples were incubated for an additional 4 h at 37°C. The purple-blue MTT formazan precipitate was dissolved in 100 μl DMSO. The absorbance was measured at 570 nm using an ELISA reader (BioTek, Model ELX800, USA).

Colony formation

Caco-2 cells were seeded into 6-well plates at a density of 1×105 cells/well in 2 ml medium. After treatment with various concentrations of PZH for 24 h, the cells were collected and diluted in fresh medium in the absence of PZH and then reseeded into 6-well plates at a density of 1×103 cells/well. Following incubation for 8 days in a 37°C humidified incubator with 5% CO2, the formed colonies were fixed with 10% formaldehyde, stained with 0.01% crystal violet and counted. Cell survival was calculated by normalizing the survival of the control cells as 100%.

Cell cycle analysis

The cell cycle analysis was carried out by flow cytometry using a fluorescence-activated cell sorting (FACS) Calibur (Becton-Dickinson, San Jose, CA, USA) and propidium iodide (PI) staining. Subsequent to treatment with various concentrations of PZH for 24 h, Caco-2 cells were collected and adjusted to a concentration of 1x106 cells/ml, and fixed in 70% ethanol at 4°C overnight. The fixed cells were washed twice with cold PBS, and then incubated for 30 min with RNase (8 μg/ml) and PI (10 μg/ml). The fluorescent signal was detected through the FL2 channel and the proportion of DNA in different phases was analyzed using ModfitLT version 3.0 (Verity Software House, Topsham).

RT-PCR analysis

Caco-2 cells were seeded into 6-well plates at a density of 1×105 cells/well in 2 ml medium and treated with various concentrations of PZH for 24 h. Total RNA was isolated with TRIzol reagent. Oligo(dT)-primed RNA (1 μg) was reverse-transcribed with SuperScript II reverse transcriptase (Promega) according to the manufacturer’s instructions. The obtained cDNA was used to determine the mRNA amount of Cyclin D1 and CDK4 by PCR. GAPDH was used as an internal control. The primer sequences used for the amplification of Cyclin D1, CDK4 and GAPDH transcripts were: Cyclin D1, forward: 5′-TGG ATG CTG GAG GTC TGC GAG GAA -3′ and reverse: 5′-GGC TTC GAT CTG CTC CTG GCA GGC-3′; CDK4, forward: 5′-CAT GTA GAC CAG GAC CTA AGC-3′ and reverse: 5′-AAC TGG CGC ATC AGA TCC TAG-3′; GAPDH, forward: 5′-CG ACC ACT TTG TCA AGC TCA-3′ and reverse: 5′-AG GGG TCT ACA TGG CAA CTG-3′. Samples were analyzed by gel electrophoresis (1.5% agarose). The DNA bands were examined using a Gel Documentation System (BioRad, Model Gel Doc 2000, USA).

Western blotting

Caco-2 cells (2.5×105 cells/well) were seeded into 25 cm2 flasks in 5 ml medium. Cells were treated with various concentrations of PZH for 24 h and then lysed with mammalian cell lysis buffer containing protease and phosphatase inhibitor cocktails. The lysates were resolved in 12% SDS-PAGE gels and electroblotted. The PVDF membranes were blocked with 5% skimmed milk and probed with primary antibodies against CyclinD1, CDK4 and β-actin (1:1,000) overnight at 4°C and then with appropriate HRP-conjugated secondary antibody followed by enhanced chemiluminescence detection.

Statistical analysis

Data were analyzed using the SPSS package for Windows (Version 11.5). Statistical analysis of the data was performed using the Student’s t-test and one-way ANOVA. Differences with P<0.05 were considered statistically significant.

Results

PZH inhibits the proliferation of Caco-2 cells

The viability of Caco-2 cells was determined by MTT assay to compare the relative number of cells in PZH-treated monolayers to untreated controls. As shown in Fig. 1A, treatment with 0.25–1 mg/ml of PZH for 24 h dose-dependently reduced cell viability by 11–35% compared to the untreated control cells (P<0.05). We also evaluated the effect of 1 mg/ml of PZH on cell viability with incubation for different periods of time. As shown in Fig. 1B, PZH treatment led to a gradual decrease in cell viability with the increase of exposure time. To verify these results, we examined the effect of PZH on Caco-2 cell survival using a colony formation assay. As shown in Fig. 2A and B, PZH treatment dose-dependently reduced the cell survival rate by 15–52% compared to the untreated control cells (P<0.05). Taken together, these data suggest that PZH inhibits Caco-2 cell proliferation in a dose- and time-dependent manner.

PZH blocks G1/S progression of Caco-2 cells

The effect of PZH on the G1 to S progression in Caco-2 cells by PI staining, followed by FACS analysis. Subsequent to treatment with 0, 0.25, 0.5 and 1 mg/ml of PZH the percentage of S-phase cells was found to be 24.0, 15.0, 10.59 and 4.11%, respectively (P<0.05) (Fig. 3), indicating that the inhibitory effect of PZH on Caco-2 cell proliferation is correlated with the arrest of G1/S cell cycle progression.

PZH regulates the expression of Cyclin D1 and CDK4 in Caco-2 cells

To explore the mechanism of the anti-proliferative activity of PZH, we performed RT-PCR and western blotting to respectively examine the mRNA and protein expression of Cyclin D1 and CDK4 in Caco-2 cells. As shown in Fig. 4, PZH treatment profoundly and dose-dependently reduced the expression of Cyclin D1 and CDK4, at both the transcriptional and translational levels.

Discussion

Drug resistance and toxicity against normal cells limit the effectiveness of current cancer chemotherapies, including those used to treat colorectal cancer (35), emphasizing the need for the development of novel cancer chemotherapies. Natural products have received a lot of attention due to their relatively few side-effects compared to modern chemotherapeutics and have been used clinically for thousands of years as important alternative remedies for various diseases, including cancer (6,7). PZH a well-known traditional Chinese formula, first prescribed 450 years ago in the Ming Dynasty, has long been used in China for cancer treatment (2224). Although it has been shown that PZH inhibits colon cancer growth via the induction of apoptosis and inhibition of tumor angiogenesis (2628), the precise mechanism of its anticancer effect remains largely unclear. Therefore, in order for PZH to be developed further as an anticancer agent, its underlying molecular mechanism of action should be elucidated.

Cancer cells are characterized by an uncontrolled proliferation (15). Therefore, inhibiting excessive proliferation of tumor cells is one of the key approaches for the development of anticancer drugs. Using MTT and colony formation analyses, we demonstrated that PZH inhibited the proliferation of human colon carcinoma Caco-2 cells, in a dose- and time-dependent manner. Eukaryotic cell proliferation is primarily regulated by the cell cycle. G1/S transition is one of the two main checkpoints of the cell cycle, responsible for initiation and completion of DNA replication (16). By using FACS analysis with PI staining, we found that PZH dose-dependently repressed the G1 to S transition in Caco-2 cells. G1/S progression is tightly regulated by the pro-proliferative Cyclin D1 and CDK4 (17). Overexpression of Cyclin D1 and CDK4 is commonly detected in various types of cancer (1821). Consistent with the inhibitory effect of PZH on G1/S transition, our data indicated that PZH treatment suppressed the mRNA and protein expression of Cyclin D1 and CDK4 in Caco-2 cells. In conclusion, the present study has demonstrated for the first time that PZH inhibited cancer cell proliferation by blocking G1 to S progression, which may be one of the mechanisms mediating its antitumor activity.

Abbreviations:

CRC

colorectal cancer;

PZH

Pien Tze Huang;

TCM

traditional Chinese medicine;

MTT

3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

Acknowledgements

This study was sponsored by the National Natural Science Foundation of China (no. 81073097), the Developmental Fund of Chen Keji Integrative Medicine (no. CKJ 2011001), and the China Postdoctoral Science Foundation (no. 2012M511437).

References

1. 

A JemalF BrayMM CenterJ FerlayE WardD FormanGlobal cancer statisticsCA Cancer J Clin616990201110.3322/caac.20107

2. 

DM GustinDE BrennerChemoprevention of colon cancer: current status and future prospectsCancer Metast Rev21323348200210.1023/A:102127122947612549770

3. 

DB LongleyWL AllenPG JohnstonDrug resistance, predictive markers and pharmacogenomics in colorectal cancerBiochim Biophys Acta1766184196200616973289

4. 

Y SunH ZhaoY GuoF LinL TangY YaoClinical study of combining chemotherapy of oxaliplatin or 5-fluorouracil/leucovorin with hydroxycamptothecine for advanced colorectal cancerClin Oncol Cancer Res6117123200910.1007/s11805-009-0117-8

5. 

G BooseH StopperGenotoxicity of several clinically used topoisomerase II inhibitorsToxicol Lett116716200010.1016/S0378-4274(00)00192-210906417

6. 

DJ NewmanGM CraggKM SnaderThe influence of natural products upon drug discoveryNat Prod Rep17215234200010.1039/a902202c10888010

7. 

M GordalizaNatural products as leads to anticancer drugsClin Transl Oncol9767776200710.1007/s12094-007-0138-918158980

8. 

JM LinYQ ChenLH WeiXZ ChenW XuZF HongTJ SferraJ PengHedyotis Diffusa Willd extract induces apoptosis via activation of the mitochondrion-dependent pathway in human colon carcinoma cellsInt J Oncol37133113382010

9. 

J PengYQ ChenJM LinQC ZhuangW XuZF HongTJ SferraPatrinia scabiosaefolia extract suppresses prolife-ration and promotes apoptosis by inhibiting STAT3 pathway in human multiple myeloma cellsMol Med Rep43133182011

10. 

JM LinLH WeiW XuZF HongXX LiuJ PengEffect of Hedyotis Diffusa Willd extract on tumor angiogenesisMol Med Rep4128312882011

11. 

QY CaiJM LinLH WeiL ZhangLL WangYZ ZhanJW ZengW XuAL ShenZF HongJ PengHedyotis diffusa Willd inhibits colorectal cancer growth in vivo via inhibition of STAT3 signaling pathwayInt J Mol Sci1361176128201210.3390/ijms13056117

12. 

LH WeiYQ ChenJM LinJY ZhaoXZ ChenW XuXX LiuTJ SferraJ PengScutellaria Barbata D. Don induces apoptosis of human colon carcinoma cell via activation of the mitochondrion-dependent pathwayJ Med Plants Res5196219702011

13. 

LH WeiJM LinW XuZF HongXX LiuTJ SferraJ PengInhibition of tumor angiogenesis by Scutellaria Barbata D. Don via suppressing proliferation, migration and tube formation of endo thelial cells and downregulation of the expression of VEGF-A in cancer cellsJ Med Plants Res5326032682011

14. 

LP ZhengYQ ChenW LinQC ZhuangXZ ChenW XuXX LiuJ PengTJ SferraSpica Prunellae extract promotes mitochondrion-dependent apoptosis in a human colon carcinoma cell lineAfr J Pharm Pharmacol5327335201110.5897/AJPP10.354

15. 

GI EvanKH VousdenProliferation, cell cycle and apoptosis in cancerNature411342348200110.1038/3507721311357141

16. 

P NurseOrdering S phase and M phase in the cell cycleCell79547550199410.1016/0092-8674(94)90539-87954820

17. 

DO MorganPrinciples of CDK regulationNature374131134199510.1038/374131a07877684

18. 

S HarakehK Abu-El-ArdatM Diab-AssafA NiedzwieckiM El-SabbanM RathEpigallocatechin-3-gallate induces apoptosis and cell cycle arrest in HTLV-1-positive and-negative leukemia cellsMed Oncol253039200810.1007/s12032-007-0036-618188712

19. 

D KesselY LuoCells in cryptophycin-induced cell-cycle arrest are susceptible to apoptosisCancer Lett1512529200010.1016/S0304-3835(99)00409-710766419

20. 

A PurohitHAM HejazL WaldenL MacCarthy-MorroghG PackhamBVL PotterMJ ReedThe effect of 2-methoxyoestrone-3-O-sulphamate on the growth of breast cancer cells and induced mammary tumoursInt J Cancer855845892000

21. 

BT ZafonteJ HulitDF AmanatullahC AlbaneseC WangE RosenA ReutensJA SparanoMP LisantiRG PestellCell-cycle dysregulation in breast cancer: breast cancer therapies targeting the cell cycleFront Biosci5D938D961200010.2741/zafonte11102317

22. 

Chinese Pharmacopoeia CommissionPharmacopoeia of the People’s Republic of China1Chinese Medical Science and Technology PressBeijing5735752010

23. 

YY XuEX YuClinical analysis of the effect of Pien Tze Huang in treatment of 42 patients with moderate or advanced liver cancerShanghai J Tradit Chin Med12451994

24. 

ZX GuTherapeutical observation of advanced colon cancerChin Tradit Patent Med15231993

25. 

CS LiuReview of Pharmacology and clinical application of Pien Tze HuangMed Pharm World764662006

26. 

JM LinLH WeiYQ ChenXX LiuZF HongTJ SferraJ PengPien Tze Huang-induced apoptosis in human colon cancer HT-29 cells is associated with regulation of the Bcl-2 family and activation of caspase 3Chin J Integr Med17685690201110.1007/s11655-011-0846-421910070

27. 

QC ZhuangF HongAL ShenLP ZhengJW ZengW LinYQ ChenTJ SferraZF HongJ PengPien Tze Huang inhibits tumor cell proliferation and promotes apoptosis via suppressing the STAT3 pathway in colorectal cancer mouseInt J Oncol4015691574201222218594

28. 

AL ShenF HongLY LiuJM LinQC ZhuangZF HongTJ SferraJ PengEffects of Pien Tze Huang on angiogenesis in vivo and in vitroChin J Integr Med18431436201210.1007/s11655-012-1121-z22821655

Related Articles

Journal Cover

October 2012
Volume 4 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shen A, Hong F, Liu L, Lin J, Wei L, Cai Q, Hong Z and Peng J: Pien Tze Huang inhibits the proliferation of human colon carcinoma cells by arresting G1/S cell cycle progression. Oncol Lett 4: 767-770, 2012
APA
Shen, A., Hong, F., Liu, L., Lin, J., Wei, L., Cai, Q. ... Peng, J. (2012). Pien Tze Huang inhibits the proliferation of human colon carcinoma cells by arresting G1/S cell cycle progression. Oncology Letters, 4, 767-770. https://doi.org/10.3892/ol.2012.811
MLA
Shen, A., Hong, F., Liu, L., Lin, J., Wei, L., Cai, Q., Hong, Z., Peng, J."Pien Tze Huang inhibits the proliferation of human colon carcinoma cells by arresting G1/S cell cycle progression". Oncology Letters 4.4 (2012): 767-770.
Chicago
Shen, A., Hong, F., Liu, L., Lin, J., Wei, L., Cai, Q., Hong, Z., Peng, J."Pien Tze Huang inhibits the proliferation of human colon carcinoma cells by arresting G1/S cell cycle progression". Oncology Letters 4, no. 4 (2012): 767-770. https://doi.org/10.3892/ol.2012.811