Isoquercitrin inhibits the progression of liver cancer in vivo and in vitro via the MAPK signalling pathway

Retraction in: /10.3892/or.2023.8510

  • Authors:
    • Guihong Huang
    • Bo Tang
    • Kun Tang
    • Xiaomin Dong
    • Jungang Deng
    • Luqin Liao
    • Zengzhen Liao
    • Hua Yang
    • Songqing He
  • View Affiliations

  • Published online on: March 20, 2014     https://doi.org/10.3892/or.2014.3099
  • Pages: 2377-2384
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Liver cancer is a malignant tumour with high morbidity and fatality rates that is common worldwide. At present, the clinical approaches to treating primary liver cancer include partial hepatectomy, systemic or local chemotherapy, radiotherapy, radiofrequency ablative surgery and liver transplantation. However, all of these approaches have shortcomings, including poor prognosis and numerous side-effects. A large number of studies have proven that many effective ingredients in traditional Chinese medicine, particularly the flavonoid compounds extracted from plants, have achieved breakthroughs in terms of enhancing the effects and reducing the toxicity of chemotherapy and radiotherapy, preventing tumour metastasis and relapse after surgery, alleviating the clinical symptoms of advanced tumours, improving the quality of life of the patient with tumours and extending patient long‑term survival. The purpose of the present study was to investigate the impact of isoquercitrin, the flavonoid from Bidens bipinnata L. extract, on the progression of liver cancer and to achieve a deeper understanding of the biological characteristics of isoquercitrin's involvement in the progression of liver cancer. In the in vitro experiments, isoquercitrin was found to strongly inhibit the proliferation of human liver cancer cells, promote the apoptosis of human liver cancer cells, and block the cell cycle in the G1 phase. Isoquercitrin activated caspase-3, -8 and -9, inhibited the expression level of ERK and p38MAPK protein phosphorylation, and promoted the phosphorylation of JNK. Additionally, isoquercitrin reduced the expression level of PKC in human liver cancer cells. In the in vivo experiments, isoquercitrin was also found to significantly inhibit the growth of transplanted tumours in nude mice. The present study confirmed that isoquercitrin could inhibit the progression of human liver cancer in vivo and in vitro, and the molecular mechanism of isoquercitrin may be closely associated with the MAPK and PKC signalling pathways.

Introduction

Liver cancer is a malignant tumour with high morbidity and fatality rates that is common worldwide (1). New cases of liver cancer account for 4% of all malignant tumours in the world each year, and the morbidity of liver cancer in China has been slowly increasing in recent years, seriously affecting the life and health of its citizens (2). With the morbidity of liver cancer in China now higher than that of gastric cancer, liver cancer currently has the highest fatality rate among malignant tumours of the digestive tract. The number of liver cancer patients in China accounts for 55% of all cases in the world, and the death toll has reached 45% of the world total (3,4). The occurrence of liver cancer is related to viral hepatitis, excessive drinking and non-alcoholic cirrhosis, all of which may increase the morbidity of liver cancer (5). Additionally, because the onset of liver cancer is hidden, most patients are already at the end-stage once a definite diagnosis is made (5,6), depriving them of the chance for radical treatment. Currently, the main clinical approaches to treating primary liver cancer include partial hepatectomy, systemic or local chemotherapy, radiotherapy, radiofrequency ablative surgery and liver transplantation (711). However, all of these approaches have shortcomings, including poor prognosis and many side-effects. Thus, there is an urgent need to search for new therapeutics with effective antitumour action for liver cancer patients.

Due to the extremely high costs for developing chemical synthetic drugs, it is quite difficult to discover an anticancer drug from chemical compounds. Additionally, chemical drugs can have many side-effects. Therefore, it has become an important part of research on anticancer drugs both at home and abroad to search for a single Chinese herb or effective ingredient from plants. As one of the key approaches to tumour treatment, traditional Chinese medicine plays an indispensable role. With decades of effort, traditional Chinese medicine has achieved breakthroughs in terms of enhancing the effect and reducing the toxicity of chemotherapy and radiotherapy, preventing tumour metastasis and relapse after surgery, alleviating the clinical symptoms of advanced tumours, improving the quality of life of patients with tumours and extending the patient’s long-term survival. Traditional Chinese medicine has become a ‘hot’ topic in cancer drug research due to its antitumour characteristics of multi-target, multi-step and multi-effect, its action in various steps in the occurrence and development of tumours, its lower toxicity and fewer toxic effects, its ability to improve immunity and its lower tendency toward drug resistance. The anticancer mechanism of Chinese medicine is also a focus in today’s medical research.

The occurrence of liver cancer is associated not only with abnormal cell proliferation and differentiation but also with abnormal apoptosis. The proliferation and apoptosis of tumour cells consist of a precise process regulated by multiple genes. Many studies both at home and abroad have demonstrated that flavonoid compounds can inhibit the proliferation of tumour cells, induce the apoptosis of tumour cells and regulate the expression of relevant genes to block the occurrence and development of tumour cells (1215). Isoquercitrin, the effective monomer from Bidens bipinnata L. extract, was used in the present study. The molecular formula of isoquercitrin is C20H21O12, with a molecular weight of 464.38 (Fig. 1 illustrates its structural formula). Isoquercitrin was used to treat human liver cancer cells to study its role in regulating the proliferation, apoptosis and cell cycle of liver cancer cells and to explore a possible antitumour signalling pathway.

Materials and methods

Cell culture, antibodies and reagents

Human liver cancer cell lines HepG2 and Hep3B were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). The cells were inoculated in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal bovine serum (FBS) (both from Gibco, USA) in a 37°C incubator containing 5% CO2 and saturated humidity. Isoquercitrin (≥98% purity) was purchased from Sigma, USA. The Annexin V-FITC apoptosis analysis kit was purchased from BD, USA. Antibodies for phospho-JNK, phospho-ERK1/2, phospho-p38MAPK and PKC were purchased from Santa Cruz Biotechnology, USA. The RT-PCR kit was purchased from Takara Bio (Japan).

Cell viability analysis

Liver cancer cells in logarithmic growth phase were digested with 0.25% trypsin, rinsed with phosphate-buffered saline (PBS), and resuspended in DMEM culture medium containing 10% FBS as single cells to be inoculated into a 96-well plate (1×104 cells/well) for overnight culture in a 5% CO2 incubator at 37°C. The following day, the medium was replaced, and isoquercitrin was added to final concentrations of 0, 100, 200, 400 and 800 μM. A blank was established in one well with culture medium only. There were 6 double-wells for each group that was cultured for 24, 48 and 72 h. The medium was replaced 4 h prior to the analysis, and MTT was added (20 μl at 0.5 mg/ml) into each well. Cells were cultured for 4 h in the incubator at 37°C. DMSO (150 μl) was added, and the OD values of each well were measured with a microplate reader at the wavelength of 490 nm.

Annexin V-FITC/PI double stain flow cytometry for apoptosis analysis

Cells in the logarithmic growth phase were inoculated in a 6-well plate, and the cell density was adjusted to 1×106 cells/well. After adherence, cells were treated with different concentrations of isoquercitrin added to the culture medium. Cells were trypsinised 48 h after the isoquercitrin treatment, and then, Annexin V-FITC (5 μl) and propidium iodide (PI) (10 μl) were added to the cell suspension, which was mixed and incubated for 15 min at room temperature in the dark before the flow cytometric analysis. This procedure was repeated with each experimental group three times.

Caspase activity assay

HepG2 cells were treated with different concentrations of isoquercitrin (0, 100, 200, 400 and 800 μM) for 48 h, collected and tested for the viability of caspase-3, -8 and -9 using a caspase activity assay kit according to the manufacturer’s instructions (Beyotime, China). Fluorescence was measured using a spectrophotometer at an excitation wavelength of 400 nm and an emission wavelength of 505 nm.

Cell cycle analysis

Cells in the logarithmic growth phase were inoculated in a 6-well plate at a density of 1×106 cells/well. After adherence, cells were treated with different concentrations of isoquercitrin added to the culture medium. Cells were then trypsinised 48 h after the isoquercitrin treatment and resuspended in solution. Cold absolute ethyl alcohol (75%) was added, and cells were fixed for >18 h at 4°C. Cells were washed twice with PBS, treated with RNase A (50 mg/l) for 30 min at 37°C, and subsequently put in an ice-bath for 2 min. Cells were stained with PI dye (50 mg/l) for 30 min at 4°C, then analysed using FACS. CellQuest software was used to analyse the distribution of all cells.

Western blot assays

Cells from all treatment conditions were collected and washed twice with chilled PBS. Lysis buffer was added, and then cells were placed on ice for 20 min and subsequently centrifuged at 13,000 rpm for 20 min at 4°C. The supernatant was collected, and total cellular protein was extracted. Protein was quantified using the BCA method. Samples were subjected to SDS-PAGE gel electrophoresis at 80 V and 4°C. Samples were electrophoresed in the stacking gel for 1 h, and then electrophoresed at 150 V for ~1.5 h until the bromophenol blue dye front reached the bottom of the separation gel, and the power was turned off. Separated lysates were then transferred to a membrane for 2 h at 100 V. Membranes were stained with Ponceau S and blocked with 5% milk via shaking at room temperature for 1 h. Primary antibodies were diluted in Tris-buffered saline Tween-20 (TBST) and incubated overnight at 4°C. Antibody dilutions were as follows: phospho-ERK1/2, 1:500; phospho-JNK, 1:800; phospho-p38, 1:800 and PKC, 1:600. Secondary antibodies were incubated via shaking at 37°C for 1 h, washed three times with TBST for 10 min each, and developed using chemiluminescence. The target protein expression levels were normalised using a ratio to β-actin levels.

RT-PCR

Total RNA was extracted from all cells and was subjected to reverse transcription following determination of purity and integrity. RNA concentration was calculated and RT-PCR reactions were performed according to the manufacturer’s instructions (Takara Bio). β-actin and PKC primers were synthesised by Invitrogen using the following sequences: β-actin forward primer 5′-AAGGAAGGCTGGAAGAGTGC-3′ and reverse primer 5′-CTGGGACGACATGGAGAAAA-3′; PKC forward primer 5′-TGAATCCTCAGTGGAATGAGT-3′ and reverse primer 5′-GGTTGCTTTCTGTCTTCTGAA-3′. The PCR reaction volume was 50 μl using the following reaction conditions: 94°C for 2 min and a 94°C denaturation for 30 sec; a 60°C annealing for 30 sec; and a 72°C extension for 30 sec for a total of 32 cycles. PCR products were electrophoresed on a 1.0% agarose gel, then scanned and analysed with a gel imaging system.

Vaccination of nude mice

The animal experimentation program was approved by the Medical Ethics Committee of Guilin Medical University. Twenty male nude mice were purchased from the Animal Experiment Center of Guilin Medical University at the age of 6 to 8 weeks and a weight of ~20 g. The mice were randomly divided into two groups of 10 mice each: the model group and isoquercitrin group. Liver cancer cells were collected in the logarithmic log phase. After washing with PBS, the cells were suspended in serum-free medium. Subsequently, 200 μl of the cell suspension (containing 2×107 cells) was injected subcutaneously into the right groin area of the mice. After tumours formed, the mice were gavaged with isoquercitrin every day and observed for the growth of tumours at 7, 14, 21 and 28 days. Mice were sacrificed by cervical vertebral dislocation 4 weeks later. Subcutaneous transplanted tumour tissue was surgically removed under aseptic conditions for index analysis.

Statistical analysis

All the experimental data were analysed with SPSS 15.0 statistical software. The measurement data are expressed as the means ± standard deviation, and the counting data are expressed as percentages. The comparison among various groups was carried out using one-way analysis of variance, and the comparison between pairs within a group was conducted using the Q test. p<0.05 was considered to indicate a statistically significant difference.

Results

Isoquercitrin inhibits the proliferation of liver cancer cells

The human liver cancer cell lines HepG2 and Hep3B were treated with various concentrations of isoquercitrin (0, 100, 200, 400 and 800 μM) for 24, 48 and 72 h. Cellular activity was assayed with MTT. Isoquercitrin inhibited the proliferation of HepG2 and Hep3B cells in a time- and concentration-dependent manner (Fig. 2). When the concentration of isoquercitrin increased from 0 to 800 μM, the A490 values in the HepG2 and Hep3B cells decreased gradually; the most significant decrease was observed at 400 μM.

Isoquercitrin induces apoptosis of liver cancer cells

To confirm whether isoquercitrin induces the apoptosis of liver cancer cells, Annexin V-FITC/PI double stain flow cytometry was used to assay for the apoptosis of HepG2 cells following treatment with various concentrations of isoquercitrin for 48 h. As the concentration of isoquercitrin gradually increased, the percentage of dead HepG2 cells increased in a concentration-dependent manner when compared with the control group (Fig. 3).

Isoquercitrin activates caspases in liver cancer cells

To confirm whether isoquercitrin induces the apoptosis of liver cancer cells, the change in the activity of caspase-3, -8 and -9 was analysed after HepG2 cells were treated with various concentrations of isoquercitrin for 48 h. The activity of caspase-3, -8 and -9 increased significantly after HepG2 cells were treated with isoquercitrin (Fig. 4). Our findings showed that isoquercitrin induced the apoptosis of HepG2 cells in a caspase family-dependent manner.

Isoquercitrin inhibits the proliferation of liver cancer cells via the MAPK signalling pathway

To explore the molecular mechanism of isoquercitrin-induced inhibition of liver cancer cell proliferation, HepG2 cells were treated with various concentrations of isoquercitrin for 48 h, and then the expression and phosphorylation levels of the MAPK pathway proteins ERK, JNK and p38MAPK were assayed by western blotting. We found that as the concentration of isoquercitrin gradually increased, the phosphorylation levels of both ERK and p38MAPK decreased, and the phosphorylation level of JNK increased (Fig. 5). Our findings showed that isoquercitrin promoted the phosphorylation of JNK, which in turn promoted the apoptosis of liver cancer cells.

Isoquercitrin inhibits the proliferation of liver cancer cells via the PKC signalling pathway

To further explore the molecular mechanism of isoquercitrin-induced inhibition of liver cancer cell proliferation, HepG2 and Hep3B cells were treated with isoquercitrin for 48 h, and then changes in PKC mRNA and protein expression were measured using RT-PCR and western blot assays, respectively. We found that, following treatment with isoquercitrin, both PKC mRNA and protein expression levels dropped significantly (Fig. 6). Our findings indicate that isoquercitrin inhibits the proliferation of liver cancer cells by downregulating PKC.

Isoquercitrin blocks the liver cancer cell cycle in the G1 phase

To investigate whether isoquercitrin regulates changes in the liver cancer cell cycle, flow cytometry was used to analyse changes in the cell cycle of HepG2 cells after they were treated with various concentrations of isoquercitrin for 48 h. It was found that as the concentration of isoquercitrin gradually increased, the percentages of HepG2 cells entering the S and the G2/M phases gradually decreased, while most cells were blocked in the G1 phase (Fig. 7). Our findings showed that the anticancer role that isoquercitrin plays in the liver may be induced by the blockade of the cell cycle.

Isoquercitrin inhibits transplanted tumour growth in nude mice

We found that the tumour volume of the isoquercitrin-treated mouse group was consistently smaller than that of the control group at every time point examined. Additionally, the weight of the tumours obtained by surgery was also significantly less than that of the control group (Fig. 8). These findings strongly indicate that isoquercitrin obviously inhibits the progression of liver cancer.

Discussion

Bidens bipinnata L. is a whole dry herb of feverfew Bidens bipinnata L. that mainly grows in the warm and humid environment of south China and is widely distributed in most areas of Guangxi Province. It has historically been used as a basic drug in the local area of Guangxi to treat such diseases as malaria, diarrhoea, dysentery, hepatitis and acute nephritis, having the effects of diaphoresis, clearing heat and toxicity, and dissipating stasis. In recent years, Bidens bipinnata L. has often been used by local people to treat hypertension, hyperlipidaemia, diabetes, and anti-hepatic fibrosis and as an antitumour agent with positive effects. Many studies have proven that the flavonoid compounds of Bidens bipinnata L. have antitumour effects (1520). Isoquercitrin is a flavonoid compound isolated from Bidens bipinnata L. Ma et al (21) and Zhong et al (22) demonstrated that the main components of the total flavone of Bidens bipinnata L. are isoquercitrin and hyperin via separation using macroporous adsorption resin and high performance liquid chromatography. Zhong et al (23) and Yuan et al (24) also studied the role that the Bidens bipinnata L. flavonoid plays in protecting mice against acute liver injury using a mouse model of acute liver injury induced by circulating tumour cells (CTCs). However, no report is available on its role in liver cancer cells. The formation of liver cancer is closely associated with abnormal proliferation and apoptosis of liver cells. To understand the effect of isoquercitrin on liver cancer cells, its role in liver cancer cell inhibition was preliminarily explored in in vivo and in vitro experiments. The possible signalling pathways involved in the inhibition of liver cancer cell growth by isoquercitrin were analysed by determining the expression levels of relevant proteins and genes to provide an experimental basis for its development and use.

There are many flavonoid drugs that can inhibit the proliferation of tumour cells. Various concentrations of isoquercitrin were administered to human liver cancer cells, and the MTT method was used to analyse its impacts on cell proliferation capacity. We found that when the concentration of isoquercitrin was higher than 100 μM, the inhibition of human liver cancer cell proliferation gradually increased as the concentration of isoquercitrin increased. The inhibition of human liver cancer cell proliferation was most obvious when the concentration of isoquercitrin was 400 μM. This means that isoquercitrin can inhibit the progression of human liver cancer in vitro in a concentration-dependent manner. In addition, we found in the in vivo experiment that the tumour formation rate of the nude mice decreased and that tumour growth was restrained following treatment with isoquercitrin. This means that isoquercitrin can inhibit the progression of human liver cancer in vivo as well.

Liver cancer is caused by the proliferation of tumour cells and the unbalanced regulation of apoptosis due to the abnormal activation of cell proliferation signals and the abnormal inhibition of cell apoptotic signals (25,26). In these experiments, the apoptosis of liver cancer cells was tested using the flow cytometry Annexin V/PI double stain method following treatment with isoquercitrin for 48 h. We found that isoquercitrin induced the apoptosis of liver cancer cells in a concentration-dependent manner. The apoptosis of cells is achieved mainly through two pathways, the death receptor pathway and the mitochondrial pathway (2729). Caspases are specific cysteine aspartic acid proteases, and they are present in an inactive zymogen state in normal cells. Once a caspase is activated, it participates in initiating and executing cell apoptosis. Caspase-9 is at a comparatively upstream stage of the cell apoptotic signal transduction process. If caspase-9 is activated, it activates the downstream effector cysteine proteases caspase-3 and -8 to trigger a caspase cascade reaction, which in turn promotes subsequent cell apoptosis signals and the initiation of apoptosis. We found that the expression of caspase-3, -8 and -9 all increased following treatment with isoquercitrin. This means that isoquercitrin induces cell apoptosis by activating caspase family proteins inside human liver cancer cells.

Tumour growth is a disorder of cell cycle regulation due to changes in multiple genes. Two key points in the cell cycle regulatory mechanism are the G1 and the S phases. The blockade of tumour cells in the G1 or the S phase is an important mechanism to resist the growth of tumour cells (30,31). In the present study, changes in the cell cycle of human liver cells were analysed by flow cytometry after they were treated with isoquercitrin to determine the mechanism by which isoquercitrin inhibits the proliferation of human liver cancer cells. We found that the percentage of tumour cells in the G1 phase increased after these cells were treated with isoquercitrin for 48 h, meaning that cells were blocked in the G1 phase. Therefore, isoquercitrin had the effect of preventing human liver cancer cells from transitioning from the G1 to the S phase and the G2/M phase.

The MAPK signalling transduction pathway is a key information transfer pathway for signals to enter the cell nucleus from the cell surface and an intersection for signal transfer between cell proliferation and differentiation. It mainly consists of extracellular regulating protein kinase (ERK), c-Jun N-terminal kinase (JNK) and p38MAPK pathways. Three pathways, ERK, JNK and p38MAPK, were found to be closely associated with many malignant tumours, including breast, ovarian cancer and non-small cell lung cancer. ERK mainly takes part in the proliferation and differentiation of cells (3235), while JNK and p38MAPK mainly take part in the stress reaction and induction of apoptosis. Additionally, JNK plays a role as a proto-oncogene in hepatocellular carcinoma (36). When JNK is activated through phosphorylation, it regulates the expression of downstream target genes and the activity of target proteins to induce apoptosis (37). When Nateri et al knocked down the c-Jun gene or changed the locus of JNK phosphorylation, intestine tumours in mice became smaller, the number of tumour cells decreased, and the lifespan of the mice was extended (38). The activation of the p38MAPK pathway plays a key role in inflammation, apoptosis, cell cycle regulation and cell differentiation. The abnormal activation of the p38MAPK pathway is closely correlated to the occurrence of tumours. Some studies indicate that p38MAPK is extensively and continuously activated in many human tumours. In addition, the p38MAPK pathway participates in all processes of tumour development and metastasis. One study proved that the use of a p38MAPK inhibitor inhibited the occurrence of some types of tumours (39). Therefore, further study concerning the key genes and proteins of the MAPK signalling pathway is warranted. We found that isoquercitrin strongly inhibited the phosphorylation of ERK and p38MAPK proteins while promoting the phosphorylation of JNK, which indicates that isoquercitrin may play a role in regulating the proliferation and apoptosis of human liver cancer cells via the MAPK signalling pathway.

Protein kinase C (PKC) plays an important role in the transmembrane signal transfer process. By catalysing Ser/Thr phosphorylation of various proteins, PKC regulates the metabolism, growth, proliferation and differentiation of various types of cells. Current studies indicate that PKC is closely related to the proliferation and attack of liver cancer cells (40). We found that isoquercitrin significantly lowered the expression level of phosphorylated PKC; thus, isoquercitrin may affect the progression of liver cancer via the PKC signalling pathway.

In summary, we found that isoquercitrin inhibited the progression of human liver cancer cells. Therefore, isoquercitrin may be a potential new antitumour drug for the treatment of liver cancer.

Acknowledgements

This study was funded by the Project of The National Natural Science Foundation of China (81260661), the Traditional Chinese Medicine Project of The Health Department of Guangxi (GZPT13-45), and The Self-initiated Project of the Health Department of Guangxi (Z2010270).

References

1 

van Meer S, de Man RA, Siersema PD and van Erpecum KJ: Surveillance for hepatocellular carcinoma in chronic liver disease: evidence and controversies. World J Gastroenterol. 19:6744–6756. 2013.PubMed/NCBI

2 

Parkin DM, Bray F, Ferlay J and Pisani P: Global cancer statistics, 2002. CA Cancer J Clin. 55:74–108. 2005. View Article : Google Scholar

3 

Liu Y, Chang CC, Marsh GM and Wu F: Population attributable risk of aflatoxin-related liver cancer: systematic review and meta-analysis. Eur J Cancer. 48:2125–2136. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Li M, Qiao C, Qin L, Zhang J and Ling C: Application of traditional Chinese medicine injection in treatment of primary liver cancer: a review. J Tradit Chin Med. 32:299–307. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Maluccio M and Covey A: Recent progress in understanding, diagnosing, and treating hepatocellular carcinoma. CA Cancer J Clin. 62:394–399. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Belghiti J and Fuks D: Liver resection and transplantation in hepatocellular carcinoma. Liver Cancer. 1:71–82. 2012. View Article : Google Scholar

7 

Eguchi S, Kanematsu T, Arii S, Omata M, Kudo M, Sakamoto M, Takayasu K, Makuuchi M, Matsuyama Y and Monden M; Liver Cancer Study Group of Japan. Recurrence-free survival more than 10 years after liver resection for hepatocellular carcinoma. Br J Surg. 98:552–557. 2011.PubMed/NCBI

8 

Huang J, Yan L, Cheng Z, Wu H, Du L, Wang J, Xu Y and Zeng Y: A randomized trial comparing radiofrequency ablation and surgical resection for HCC conforming to the Milan criteria. Ann Surg. 252:903–912. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Wang W, Shi J and Xie WF: Transarterial chemoembolization in combination with percutaneous ablation therapy in unresectable hepatocellular carcinoma: a meta-analysis. Liver Int. 30:741–749. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Marelli L, Stigliano R, Triantos C, Senzolo M, Cholongitas E, Davies N, Yu D, Meyer T, Patch DW and Burroughs AK: Treatment outcomes for hepatocellular carcinoma using chemoembolization in combination with other therapies. Cancer Treat Rev. 32:594–606. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Roomi MW, Roomi NW, Kalinovsky T, Niedzwiecki A and Rath M: Micronutrient synergy in the fight against hepatocellular carcinoma. Cancers. 4:323–339. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Ghosh A, Ghosh D, Sarkar S, Mandal AK, Thakur Choudhury S and Das N: Anticarcinogenic activity of nanoencapsulated quercetin in combating diethylnitrosamine-induced hepatocarcinoma in rats. Eur J Cancer Prev. 21:32–41. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Khan MS, Halagowder D and Devaraj SN: Methylated chrysin induces co-ordinated attenuation of the canonical Wnt and NF-kB signaling pathway and upregulates apoptotic gene expression in the early hepatocarcinogenesis rat model. Chem Biol Interact. 193:12–21. 2011. View Article : Google Scholar

14 

Liu H, Dong A, Gao C, Tan C, Xie Z, Zu X, Qu L and Jiang Y: New synthetic flavone derivatives induce apoptosis of hepatocarcinoma cells. Bioorg Med Chem. 18:6322–6328. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Ullmannova V and Popescu NC: Inhibition of cell proliferation, induction of apoptosis, reactivation of DLC1, and modulation of other gene expression by dietary flavone in breast cancer cell lines. Cancer Detect Prev. 31:110–118. 2007.PubMed/NCBI

16 

Yang QH, Yang J, Liu GZ, Wang L, Zhu TC, Gao HL and Kou XG: Study on in vitro anti-tumor activity of Bidens bipinnata L. extract. Afr J Tradit Complement Altern Med. 10:543–549. 2013.PubMed/NCBI

17 

Wu J, Wan Z, Yi J, Wu Y, Peng W and Wu J: Investigation of the extracts from Bidens pilosa Linn. var radiata Sch Bip for antioxidant activities and cytotoxicity against human tumor cells. J Nat Med. 67:17–26. 2013.

18 

Kumari P, Misra K, Sisodia BS, Faridi U, Srivastava S, Luqman S, Darokar MP, Negi AS, Gupta MM, Singh SC and Kumar JK: A promising anticancer and antimalarial component from the leaves of Bidens pilosa. Planta Med. 75:59–61. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Kviecinski MR, Felipe KB, Schoenfelder T, de Lemos Wiese LP, Rossi MH, Gonçalez E, Felicio JD, Filho DW and Pedrosa RC: Study of the antitumor potential of Bidens pilosa (Asteraceae) used in Brazilian folk medicine. J Ethnopharmacol. 117:69–75. 2008.PubMed/NCBI

20 

Ong PL, Weng BC, Lu FJ, Lin ML, Chang TT, Hung RP and Chen CH: The anticancer effect of protein-extract from Bidens alba in human colorectal carcinoma SW480 cells via the reactive oxidative species- and glutathione depletion-dependent apoptosis. Food Chem Toxicol. 46:1535–1547. 2008.PubMed/NCBI

21 

Ma TT, Xie J, Zhang QL, Xu H, Li J and Chen FH: Analysis of fingerprint and bioactive components of Bidens biternata by HPLC. Zhong Yao Cai. 35:892–896. 2012.(In Chinese).

22 

Zhong MM, Chen FH, Yuan LP, Wang XH and Wu FR: Study on the property of adsorption and separation of the macroporous resins for total flavonoids of Bidens bipinnata L. Zhong Yao Cai. 30:338–341. 2007.(In Chinese).

23 

Zhong MM, Chen FH, Yuan LP, Wang XH, Wu FR, Yuan FL and Cheng WM: Protective effect of total flavonoids from Bidens bipinnata L. against carbon tetrachloride-induced liver injury in mice. J Pharm Pharmacol. 59:1017–1025. 2007.PubMed/NCBI

24 

Yuan LP, Chen FH, Ling L, Bo H, Chen ZW, Li F, Zhong MM and Xia LJ: Protective effects of total flavonoids of Bidens bipinnata L. against carbon tetrachloride-induced liver fibrosis in rats. J Pharm Pharmacol. 60:1393–1402. 2008.PubMed/NCBI

25 

Fabregat I, Roncero C and Fernández M: Survival and apoptosis: a dysregulated balance in liver cancer. Liver Int. 27:155–162. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Koschny R, Brost S, Hinz U, Sykora J, Batke EM, Singer S, Breuhahn K, Stremmel W, Walczak H, Schemmer P, Schirmacher P and Ganten TM: Cytosolic and nuclear caspase-8 have opposite impact on survival after liver resection for hepatocellular carcinoma. BMC Cancer. 13:5322013. View Article : Google Scholar : PubMed/NCBI

27 

Kumar S and Vaux DL: Apoptosis. A cinderella caspase takes center stage. Science. 297:1290–1291. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Dewson G and Kluck RM: Mechanisms by which Bak and Bax permeabilise mitochondria during apoptosis. J Cell Sci. 122:2801–2808. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Fombonne J, Bissey PA, Guix C, Sadoul R, Thibert C and Mehlen P: Patched dependence receptor triggers apoptosis through ubiquitination of caspase-9. Proc Natl Acad Sci USA. 109:10510–10515. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Rew DA and Wilson GD: Cell production rates in human tissues and tumours and their significance. Part II: clinical data. Eur J Surg Oncol. 26:405–417. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Kazi A and Dou QP: Cell cycle and drug sensitivity. Methods Mol Med. 111:33–42. 2005.

32 

Fecher LA, Amaravadi RK and Flaherty KT: The MAPK pathway in melanoma. Curr Opin Oncol. 20:183–189. 2008. View Article : Google Scholar

33 

Junttila MR, Li SP and Westermarck J: Phosphatase-mediated crosstalk between MAPK signaling pathways in the regulation of cell survival. FASEB J. 22:954–965. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Roberts PJ and Der CJ: Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 26:3291–3310. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Kohno M and Pouyssegur J: Targeting the ERK signaling pathway in cancer therapy. Ann Med. 38:200–211. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Das M, Garlick DS, Greiner DL and Davis RJ: The role of JNK in the development of hepatocellular carcinoma. Genes Dev. 25:634–645. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Dhanasekaran DN and Reddy EP: JNK signaling in apoptosis. Oncogene. 27:6245–6251. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Nateri AS, Spencer-Dene B and Behrens A: Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development. Nature. 437:281–285. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Marengo B, De Ciucis CG, Ricciarelli R, Furfaro AL, Colla R, Canepa E, Traverso N, Marinari UM, Pronzato MA and Domenicotti C: p38MAPK inhibition: a new combined approach to reduce neuroblastoma resistance under etoposide treatment. Cell Death Dis. 4:e5892013. View Article : Google Scholar : PubMed/NCBI

40 

Guo K, Liu Y, Zhou H, Dai Z, Zhang J, Sun R, Chen J, Sun Q, Lu W, Kang X and Chen P: Involvement of protein kinase C β-extracellular signal-regulating kinase 1/2/p38 mitogen-activated protein kinase-heat shock protein 27 activation in hepatocellular carcinoma cell motility and invasion. Cancer Sci. 99:486–496. 2008.

Related Articles

Journal Cover

May-2014
Volume 31 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
Huang G, Tang B, Tang K, Dong X, Deng J, Liao L, Liao Z, Yang H and He S: Isoquercitrin inhibits the progression of liver cancer in vivo and in vitro via the MAPK signalling pathway Retraction in /10.3892/or.2023.8510. Oncol Rep 31: 2377-2384, 2014
APA
Huang, G., Tang, B., Tang, K., Dong, X., Deng, J., Liao, L. ... He, S. (2014). Isoquercitrin inhibits the progression of liver cancer in vivo and in vitro via the MAPK signalling pathway Retraction in /10.3892/or.2023.8510. Oncology Reports, 31, 2377-2384. https://doi.org/10.3892/or.2014.3099
MLA
Huang, G., Tang, B., Tang, K., Dong, X., Deng, J., Liao, L., Liao, Z., Yang, H., He, S."Isoquercitrin inhibits the progression of liver cancer in vivo and in vitro via the MAPK signalling pathway Retraction in /10.3892/or.2023.8510". Oncology Reports 31.5 (2014): 2377-2384.
Chicago
Huang, G., Tang, B., Tang, K., Dong, X., Deng, J., Liao, L., Liao, Z., Yang, H., He, S."Isoquercitrin inhibits the progression of liver cancer in vivo and in vitro via the MAPK signalling pathway Retraction in /10.3892/or.2023.8510". Oncology Reports 31, no. 5 (2014): 2377-2384. https://doi.org/10.3892/or.2014.3099