Isoquercitrin inhibits the progression of pancreatic cancer in vivo and in vitro by regulating opioid receptors and the mitogen-activated protein kinase signalling pathway

Retraction in: /10.3892/or.2023.8505

  • Authors:
    • Quan Chen
    • Ping Li
    • Yong Xu
    • Yang Li
    • Bo Tang
  • View Affiliations

  • Published online on: November 26, 2014     https://doi.org/10.3892/or.2014.3626
  • Pages: 840-848
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pancreatic cancer is a common malignant tumour that affects individuals worldwide. In recent years, the incidence and mortality rates of pancreatic cancer have continuously increased. Currently, the primary clinical treatment methods for pancreatic cancer include surgical resection, chemotherapy and radiotherapy. However, these treatment methods rarely produce satisfactory therapeutic outcomes. Extensive research has also proven that the effective components of several traditional Chinese medicines, particularly flavonoids extracted from plants, have significant antitumour effects. Isoquercitrin, which is one of the flavonoids found in Bidens pilosa extracts, has a significant antitumour effect. However, the antitumour effect of isoquercitrin and its mechanism of action remain unclear. The objective of the present study was to investigate the effect of isoquercitrin on the progression of pancreatic cancer and to further understand the biological characteristics of the participation of isoquercitrin in the progression of pancreatic cancer. In vitro, we found that a therapeutic dose of isoquercitrin significantly inhibited proliferation, promoted apoptosis and induced cell cycle arrest within the G1 phase in pancreatic cancer cells. Isoquercitrin activated caspase-3, -8 and -9 and reduced the mitochondrial membrane potential. In addition, isoquercitrin inhibited the expression level of the δ opioid receptor; however, isoquercitrin had no effect on the κ and µ opioid receptors. Furthermore, isoquercitrin inhibited extracellular signal-regulated kinase (ERK) phosphorylation and promoted c-Jun N-terminal kinase (JNK) phosphorylation. In vivo, we found that a therapeutic dose of isoquercitrin significantly inhibited xenograft growth in nude mice. In summary, the present study demonstrated that isoquercitrin inhibits human pancreatic cancer progression in vivo and in vitro and that its molecular mechanism may be closely related to opioid receptors and to the activation of the mitogen-activated protein kinase (MAPK) signalling pathway.

Introduction

Pancreatic cancer is a common malignant tumour of the digestive system. The incidence and mortality rates of pancreatic cancer are extremely high (1,2). In addition, pancreatic cancer is the fourth most common cause for cancer-related deaths, and the 5-year overall survival rate is less than 2% (3). In recent years, the incidence rate of pancreatic cancer has gradually increased; pancreatic cancer severely affects the lives and health of individuals. Currently, the primary treatment method for pancreatic cancer is surgical resection. However, only a small number of patients (10–20%) qualify for surgical treatment due to the difficulty in early diagnosis of this cancer (46). In addition, the results from chemotherapy and radiotherapy are often unsatisfactory due to the ability of tumour cells to evade cell death (7,8). Therefore, the search for new therapeutic drugs with effective anticancer effects is extremely urgent.

Traditional Chinese medicines have multi-target, multistage and multi-effect antitumour effects. Traditional Chinese medicines can act on various stages of tumourigenesis and tumour development. In addition, traditional Chinese medicines have minor toxic side-effects and can improve body immunity; tumour cells rarely become resistant to traditional Chinese medicines. Therefore, traditional Chinese medicines have become a focus of antitumour drug studies. Thus far, extensive research has proven that flavonoids achieve antitumour effects by inhibiting the proliferation of some tumour cells, inducing the apoptosis of tumour cells, and regulating the expression levels of related oncogenes and tumour-suppressor genes (914). Isoquercitrin is the effective monomer component of Bidens pilosa extracts. The molecular formula of isoquercitrin is C20H21O12, and the molecular weight of isoquercitrin is 464.38 (15,16). Previous studies have shown that isoquercitrin can inhibit the proliferation of various types of human tumour cells, indicating that this compound has a potential anticancer effect. In addition, the anticancer effects of isoquercitrin on liver cancer and on glioblastoma have been proven (1720). However, the antitumour effect of isoquercitrin on pancreatic cancer cells and the mechanisms of isoquercitrin antitumour action remain unclear.

The development of pancreatic cancer is related to not only the abnormal proliferation and differentiation of cells but also abnormal apoptosis. The proliferation and apoptosis of tumour cells are precise, genetically regulated processes. Previous studies have shown that the mitogen-activated protein kinase (MAPK) signal transduction pathway regulates pancreatic cancer cell proliferation and apoptosis (21,22). However, pancreatic cancer development and progression are also closely related to opioid receptor pathways (23,24). In the present study, we used the effective monomer component of Bidens pilosa extract, isoquercitrin, for research. We used isoquercitrin to interfere with pancreatic cancer cells, studied the regulating effect of isoquercitrin on the proliferation, apoptosis and cell cycle of pancreatic cancer cells, and determined the possible molecular mechanism of the antitumour effect of isoquercitrin.

Materials and methods

Cell culture, drugs and antibodies

The human pancreatic cancer cell lines BxPC-3 and AsPC-1 were both purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). The aforementioned cells were cultured in a Roswell Park Memorial Institute (RPMI)-1640 medium containing 10% foetal bovine serum (FBS) in an incubator with saturated humidity at 37°C with 5% CO2. Isoquercitrin (≥98% purity) was purchased from Sigma-Aldrich (St. Louis, MO, USA). Phosphorylated c-Jun N-terminal kinase (JNK), phosphorylated extracellular-signal-regulated kinase (ERK)1/2, and phosphorylated p38MAPK antibodies as well as δ, μ and κ opioid receptor (DOR, MOR and KOR) antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA).

The 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) assay was used for assessing cell viability

Pancreatic cancer cells in logarithmic growth phase were selected. Dulbecco’s modified Eagle’s medium (DMEM) containing 10% FBS was used for single cell suspensions. The cells were cultured in a 96-well cell culture plate (1×104 cells/well). Then, the cell culture plate was placed in an incubator overnight. Different concentrations (0, 50, 100, 200 and 400 μM) of isoquercitrin were added, with 6 replicates in each group. The cells were cultured for 24, 48 and 72 h. MTT (20 μl) (0.5 mg/ml) was added to each well, and the cell culture plate was placed in an incubator at 37°C for another 4 h. Finally, 150 μl of dimethyl sulphoxide (DMSO) was added. A microplate reader was used to measure the optical density (OD) value (A490 nm) of each well at 490 nm.

Flow cytometry was used to analyse the cell cycle

The cells were cultured in a 6-well cell culture plate at a seeding density of 1×106 cells/well. After the cells were treated with different doses of isoquercitrin for 48 h, the cells were trypsinised, and a single cell suspension was prepared. Subsequently, 70% cold anhydrous ethanol was added, and the cells were fixed at 4°C overnight. After the cells were washed with phosphate-buffered saline (PBS), the cells were treated with 50 mg/l of ribonuclease A (RNase A) at 37°C for 30 min, after which the cells were placed in an ice bath for 2 min. Next, the cells were stained with 50 mg/l propidium iodide (PI) without light exposure for 30 min at 4°C. Then, the cells were examined using a flow cytometer, and CellQuest software was used to analyse the cell cycle distribution of cells in each group.

Flow cytometry was used for assessing cell apoptosis

The cell density was adjusted to 1×106 cells/well. After treatment with different doses of isoquercitrin for 48 h, the cells were trypsinised, and a single cell suspension was prepared. Five microlitres of Annexin V-fluorescein isothiocyante (FITC) and 10 μl of PI were successively added to the cell suspension, mixed well and incubated for 20 min at room temperature without light exposure. Finally, a flow cytometer was used to assess cell apoptosis.

Reverse transcription-polymerase chain reaction (RT-PCR) analysis

Total ribonucleic acid (RNA) was extracted from the cells in each group. After purity and integrity testing, the total RNA was reverse transcribed. After the RNA concentration was calculated, an RT-PCR kit (Takara Biotechnology Co., Ltd., Dailan, China) was used to conduct the RT-PCR according to the manufacturer’s instructions. The primers used in the experiment were synthesised by Invitrogen (Carlsbad, CA, USA). Table I lists the primer sequences. The PCR reaction volume was 50 μl. The reaction conditions were as follows: 94°C for 2 min; denaturing at 94°C for 2 min, annealing at 60°C for 30 sec, and elongation at 72°C for 30 sec with 32 cycles in total. The obtained PCR products were verified using 1.0% agarose gel electrophoresis. In addition, a gel imaging system was used for scanning analysis.

Table I

Primers sequences for DOR, MOR, KOR and β-actin.

Table I

Primers sequences for DOR, MOR, KOR and β-actin.

PrimersForwardReverse
DOR 5′-ACCAAGATCTGCGTGTTCCT-3′ 5′-CGATGACGAAGATGTGGATG-3′
MOR 5′-TCTGGCTCCAAAGAAAAGGA-3′ 5′-CAATGCAGAAGTGCCAAGAA-3′
KOR 5′-CGTCTCAAGAGCGTCCG-3′ 5′-TATGTGAATGGGAGTCCAGC-3′
β-actin 5′-AAGGAAGGCTGGAAGAGTGC-3′ 5′-CTGGGACGACATGGAGAAAA-3′

[i] DOR, δ opioid receptor; MOR, μ opioid receptor; KOR, κ opioid receptor.

Western blot analysis

Each group of tumour cells was collected, and protein was extracted. The bicinchoninic acid assay (BCA) was used for protein quantification. After the protein sample of each group was loaded, electrophoresis was conducted until the bromophenol blue reached the bottom of the separation gel. Then, the machine was powered off. The proteins were transferred to a membrane at 100 V for 2 h. The membrane was blocked with 5% non-fat powdered milk at room temperature with shaking for 1 h. The primary antibody, which was diluted with Tris-buffered saline with Tween-20 (TBST) solution, was added to the membrane and incubated at 4°C overnight. The dilution ratios for the primary antibodies were as follows: phosphorylated ERK1/2 (1:800), phosphorylated JNK (1:800), phosphorylated p38 (1:800), DOR (1:800), MOR (1:800) and KOR (1:800). The secondary antibody was added to the membrane and incubated at 37°C for 1 h. The membrane was shaken and washed 3 times (15 min each time) with a TBST solution. Images were acquired using darkroom development techniques for chemiluminescence. The results were analysed after the films were developed. The grey level ratio of the target protein to β-actin was used to represent the protein expression level.

Inoculation of nude mice

The protocol for the animal experiment was approved by the Medical Ethics Committee of Guilin Medical University. Nude mice were purchased from the Animal Experiment Center of Guilin Medical University, Guilin, Guangxi, China. In total, 40 male nude mice (6–8 weeks old, ~20 g) were used. The nude mice were randomly divided into the control and the isoquercitrin group, with 20 mice in each group. After tumour formation, isoquercitrin was administered to the mice in the isoquercitrin group by intragastric administration every day, and tumour growth was observed and measured on the 7, 14, 21 and 28 days. Four weeks later, the nude mice were sacrificed by cervical vertebra dislocation. Under aseptic conditions, subcutaneous xenograft tissue was excised for index determination.

Statistical analysis

Statistical Package for the Social Sciences (SPSS) 18.0 software was used for the statistical analysis. The measurement data are presented as the means ± standard deviation, and the enumeration data are expressed as percentages. One-way analysis of variance was used for the comparison between the groups. The Q-test was used for multiple comparisons. The results were considered to indicate a statistically significant result when p<0.05.

Results

Isoquercitrin inhibits pancreatic cancer cell proliferation

Different doses (0, 50, 100, 200 and 400 μM) of isoquercitrin were used to treat BxPC-3 and AsPC-1 cells for 24, 48 and 72 h. The MTT assay was used to assess cell viability. We found that isoquercitrin had varying levels of inhibition on the proliferation of BxPC-3 and AsPC-1 cells and that this inhibitory effect was time- and dose-dependent. As the concentration of isoquercitrin increased, the A490 nm values of the BxPC-3 and AsPC-1 cells gradually decreased; the decrease in BxPC-3 and AsPC-1 cells was the most prominent when the isoquercitrin concentration was 200 μM (Fig. 1).

Isoquercitrin induces pancreatic cancer cell apoptosis

To understand the mechanism of inhibition of pancreatic cancer cells by isoquercitrin, we used a therapeutic dose of isoquercitrin to treat BxPC-3 and AsPC-1 cells. Forty-eight hours later, Annexin V-FITC/PI staining flow cytometry was used to assess cell apoptosis. Compared with the control group, we found that the number of apoptotic cells increased significantly in the isoquercitrin group (p<0.05) (Fig. 2A). In addition, we also found that a therapeutic dose of isoquercitrin could significantly increase the activities of caspase-3, -8 and -9 (Fig. 2B). The above results indicated that isoquercitrin induces caspase family-dependent apoptosis of pancreatic cancer cells.

Isoquercitrin induces cell cycle arrest in pancreatic cancer cells

To further investigate the mechanism of inhibition of pancreatic cancer cell proliferation by isoquercitrin, we used a therapeutic dose of isoquercitrin to treat BxPC-3 and AsPC-1 cells. Forty-eight hours later, flow cytometry was used to determine the cell cycle changes. We found that a therapeutic dose of isoquercitrin could reduce the numbers of BxPC-3 and AsPC-1 cells that entered the S and the G2/M phase; the majority of the cells were arrested within the G1 phase (Fig. 3A). In addition, we also found that the levels of cyclin-dependent kinase (CDK)4, CDK6 and cyclin D1, which regulate the cell cycle, were significantly decreased (Fig. 3B), indicating that the antiproliferation effect of isoquercitrin on pancreatic cancer cells may be due to cell cycle arrest.

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

To elucidate the molecular mechanism of inhibition of pancreatic cancer cell proliferation by isoquercitrin, we used different doses of isoquercitrin to treat the BxPC-3 cells. Forty-eight hours later, western blot analysis was conducted to analyse the protein and phosphorylation levels of ERK, JNK and p38MAPK. With increasing doses of isoquercitrin, we found that the ERK phosphorylation level was gradually decreased, whereas the JNK phosphorylation level was gradually increased; however, no significant change in the p38MAPK phosphorylation level was observed (Fig. 4). Our results indicate that isoquercitrin could inhibit ERK phosphorylation and promote JNK phosphorylation, which in turn, induced pancreatic cancer cell apoptosis.

Isoquercitrin inhibits pancreatic cancer cell proliferation via regulation of the opioid receptor signaling pathway

To further elucidate the molecular mechanism of the inhibition of pancreatic cancer cell proliferation by isoquercitrin, we used different doses of isoquercitrin to treat the BxPC-3 and AsPC-1 cells. Forty-eight hours later, RT-PCR and western blot analysis were used to evaluate the messenger RNA (mRNA) and protein levels, respectively, of the μ, δ and κ opioid receptors. First, we found that the δ opioid receptor was expressed in pancreatic cancer cells, whereas the μ and κ opioid receptors were not expressed (Fig. 5A). After the cells were treated with a therapeutic dose of isoquercitrin, the gene and protein expression levels of the δ opioid receptor both significantly decreased compared with the control group (p<0.05) (Fig. 5B). However, after we added the specific active δ opioid receptor, the inhibitory effect of isoquercitrin on tumour cell proliferation was significantly reduced (p<0.05) (Fig. 5C). The above results demonstrated that isoquercitrin may inhibit pancreatic cancer cell proliferation via downregulation of the δ opioid receptor.

Isoquercitrin inhibits the growth of xenografts in nude mice

The nude mice were routinely feed, and xenografts were successfully transplanted. At all of the time points, we found that the mean tumour volume of the isoquercitrin group was significantly smaller than that of the control group (p<0.05). In addition, we also found that the mean weight of the surgically removed tumours from the mice in the isoquercitrin group was significantly less than that of the surgically removed tumours from the mice in the control group (p<0.05) (Fig. 6). The above results indicated that isoquercitrin could also significantly inhibit pancreatic cancer progression in vivo.

Discussion

Since the discovery of flavonoids from Bidens pilosa, studies have focused on using these flavonoids to treat hypertension, hyperlipidaemia, diabetes and hepatic fibrosis. Bidens pilosa is the dry whole grass of composite Bidens pilosa. Bidens pilosa primarily grows in the warm and moist environment of southern China and has a relatively long history of being used for treating diseases, such as malaria, diarrhoea, dysentery and hepatitis (2528). Recently, extensive studies have found that flavonoids from Bidens pilosa have antitumour effects (2932). This discovery indicates that in addition to the aforementioned functions, flavonoids from Bidens pilosa also play an important role in antitumour activity.

Isoquercitrin is one of the flavonoids present in Bidens pilosa extracts (33). In recent years, studies have shown that isoquercitrin has anti-inflammatory, anti-allergic, antioxidant and anti-injury effects (3437); furthermore, studies on flavonoids have entered a new stage of research. Research has shown that isoquercitrin can inhibit the proliferation of some tumour cells, such as liver cancer and glioblastoma cells (1720). However, few reports have examined the effect of isoquercitrin on pancreatic cancer and its mechanism. The development and progression of pancreatic cancer are closely related to abnormal cellular proliferation and apoptosis. To elucidate the effect of isoquercitrin on pancreatic cancer, in vivo and in vitro experiments were conducted in the present study to determine the inhibitory effect of isoquercitrin on pancreatic cancer progression. In addition, the expression levels of related proteins and genes were examined to analyse the possible signalling pathways through which isoquercitrin inhibited the growth of pancreatic cancer cells. The results from the present study provide experimental bases for the utilisation of isoquercitrin.

Numerous previous studies have shown that the monomers of many plant extracts can inhibit the proliferation of various types of tumour cells. This phenomenon is more common among flavonoid extracts (3840). In the present study, we found that the proliferative ability of tumour cells was significantly affected when different doses of isoquercitrin were used to treat pancreatic cancer cells. With increasing doses of isoquercitrin, the proliferative ability of the pancreatic cancer cells was gradually decreased. The proliferative ability of pancreatic cancer cells was decreased the most when the isoquercitrin dose was 200 μM. The above result indicates that isoquercitrin had a significant, dose-dependent, inhibitory effect on pancreatic cancer progression in vitro. In addition, we also conducted in vivo experiments in which isoquercitrin was used to inhibit pancreatic cancer progression. After treatment with a therapeutic dose of isoquercitrin, we found that the tumour formation rate of xenografts in nude mice was decreased significantly and that tumour growth was inhibited, indicating that isoquercitrin also inhibits pancreatic cancer progression in vivo. The abovementioned experimental results are consistent with the tumour-inhibiting effects of the monomer components of most flavonoids.

Previous studies have shown that the carcinogenesis and progression of pancreatic cancer are closely related to the maladjustment of the regulatory mechanism of cancer cell proliferation and apoptosis (41). In addition, tumour development and progression are closely related to cell cycle progression. Research has shown that tumour cell arrest at the G1 or S phase is an extremely important molecular mechanism for antagonising tumour progression (42). Our results indicated that the number of apoptotic pancreatic cancer cells increased significantly and in a dose-dependent manner after treatment with a therapeutic dose of isoquercitrin for 48 h. In addition, we also found that the protein expression levels of caspase-3, -8 and -9 were significantly increased in the tumour cells. Extensive research has proven that the occurrence of apoptosis is primarily dependent on the death receptor pathway and on the mitochondrial pathway (43). Caspase family members participate throughout the initiation and execution processes of apoptosis. After upstream caspase-9 is activated, caspase-9 then activates downstream caspase-3 and -8 to further initiate the caspase cascade reaction, which then begins apoptosis (44). Our above mentioned research results demonstrated that isoquercitrin-induced pancreatic cancer cell apoptosis may occur by activating the caspase family. Furthermore, the G1 phase percentage of pancreatic cancer cells was increased significantly, and cell proliferation was inhibited after treatment with a therapeutic dose of isoquercitrin for 48 h in the present study, indicating that isoquercitrin had an inhibitory effect on pancreatic cancer cells transitioning from G1 to S phase, which is consistent with the results from a study conducted by Huang et al (17).

The δ opioid receptor is the primary member of the opioid receptor superfamily. It has been reported that the δ opioid receptor is widely existent in various types of malignant tumour tissues and is closely related to the survival and proliferation of tumour cells (45). However, no valid evidence exists to determine whether the δ opioid receptor acts in the same manner on pancreatic cancer. We found that the δ opioid receptor was expressed in pancreatic cancer cells. However, we found that the μ and κ opioid receptors were not expressed in pancreatic cancer cells. In addition, 48 h after treatment with a therapeutic dose of isoquercitrin, the expression level of the δ opioid receptor in pancreatic cancer cells was significantly decreased, and cell proliferation was inhibited. However, when the δ opioid receptor was activated simultaneously when isoquercitrin was used to treat the pancreatic cancer cells, the inhibitory effect of isoquercitrin on tumour cell proliferation was significantly weakened or disappeared. The above result strongly indicates that the functional status of the δ opioid receptor plays a key role in the inhibition of pancreatic cancer development and progression by isoquercitrin. Further studies are required to understand its molecular mechanism.

The MAPK signalling pathway is one of the important signal transduction systems in organisms. Extensive research has proven that the MAPK signalling pathway is an important information transmission pathway from the cell surface to the cell nucleus and is the converging point of multiple signalling pathways (cell proliferation, cell differentiation) (46,47). The MAPK signalling pathway primarily includes 3 classical signalling pathways, ERK, p38MAPK and JNK. Thus far, research has shown that these 3 signalling pathways, ERK, p38MAPK and JNK, all participate in regulating the development and progression of various types of malignant tumours. A study conducted by Robbs et al demonstrated that the ERK signalling pathway participates in regulating the proliferation and differentiation of various types of tumour cells (48). The present study demonstrated that a therapeutic dose of isoquercitrin significantly inhibitws ERK phosphorylation, which in turn, affected pancreatic cancer progression, which is consistent with the results from a study conducted by Robbs et al (48). Extensive research has proven that the JNK and p38MAPK signalling pathways primarily participate in mediating cellular apoptosis; after cytoplasmic JNK is phosphorylated, this protein regulates the downstream target protein or the activity of the target protein to mediate cellular apoptosis (49). In addition, downregulation or knockout of the c-Jun gene or altering JNK phosphorylation sites can significantly inhibit tumour progression and extend survival time (50). The results of the present study demonstrated that the phosphorylation level of JNK inside of tumour cells increased significantly and that the apoptosis of tumour cells occurred after treatment with a therapeutic dose of isoquercitrin, which further proves the aforementioned theory. Previous studies have proven that p38MAPK is generally continuously activated in many types of tumours; abnormal activation of the p38MAPK pathway is closely related to the development and progression of these tumours. Existing research has shown that inhibiting the p38MAPK pathway can have an inhibitory effect on the development of various types of malignant tumours (51,52). However, we found that a therapeutic dose of isoquercitrin could not alter the p38MAPK phosphorylation level, which may be related to the fact that pancreatic cancer and other organ tumours have specific differences and that p38MAPK expression may have multiple subtypes in pancreatic cancer. The above mentioned issues warrant further study.

In summary, we found that isoquercitrin had an inhibitory effect on pancreatic cancer progression in vivo and in vitro. In addition, the molecular mechanism of the inhibitory effect of isoquercitrin may be closely related to opioid receptors and to the activation of the MAPK signalling pathway. Therefore, isoquercitrin may soon become a new drug target in the clinical treatment of pancreatic cancer, and the present study provides important theoretical bases for searching for new types of antitumour drugs.

Acknowledgements

The present study was financially supported by the National Natural Science Foundation of China (81360367), the Special Project for Chinese Medicine Technology of the Guangxi Health Commission (GZPT13-45), the Key Science and Technology Project of Higher Education Institutions in Guangxi (2013ZD046), the Key Molecular Medicine Laboratory Construction Project for Liver Injury and Repair of Guangxi (SYS2013009), and the Self-funded Subject of the Guangxi Health Commission (Z2013464).

References

1 

Roy R and Maraveyas A: Chemoradiation in pancreatic adenocarcinoma: a literature review. Oncologist. 15:259–269. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Mian OY, Ram AN, Tuli R and Herman JM: Management options in locally advanced pancreatic cancer. Curr Oncol Rep. 16:3882014. View Article : Google Scholar : PubMed/NCBI

3 

Jemal A, Siegel R, Ward E, Murray T, Xu J and Thun MJ: Cancer statistics, 2007. CA Cancer J Clin. 57:43–66. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Lockhart AC, Rothenberg ML and Berlin JD: Treatment for pancreatic cancer: current therapy and continued progress. Gastroenterology. 128:1642–1654. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Wray CJ, Ahmad SA, Matthews JB and Lowy AM: Surgery for pancreatic cancer: recent controversies and current practice. Gastroenterology. 128:1626–1641. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Gudjonsson B: Pancreatic cancer: survival errors and evidence. Eur J Gastroenterol Hepatol. 21:1379–1382. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Fulda S: Tumor resistance to apoptosis. Int J Cancer. 124:511–515. 2009. View Article : Google Scholar

8 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Tang J, Li N, Dai H and Wang K: Chemical constituents from seeds of Alpinia katsumadai, inhibition on NF-κB activation and anti-tumor effect. Zhongguo Zhong Yao Za Zhi. 35:1710–1714. 2010.(In Chinese). PubMed/NCBI

10 

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

11 

Vogel S, Ohmayer S, Brunner G and Heilmann J: Natural and non-natural prenylated chalcones: synthesis, cytotoxicity and anti-oxidative activity. Bioorg Med Chem. 16:4286–4293. 2008. View Article : Google Scholar : PubMed/NCBI

12 

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

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 : PubMed/NCBI

14 

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. View Article : Google Scholar : PubMed/NCBI

15 

Valentová K, Vrba J, Bancířová M, Ulrichová J and Křen V: Isoquercitrin: pharmacology, toxicology, and metabolism. Food Chem Toxicol. 68:267–282. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Liu Z, Zhang A, Guo Y and Dong C: Electrochemical sensor for ultrasensitive determination of isoquercitrin and baicalin based on DM-β-cyclodextrin functionalized graphene nanosheets. Biosens Bioelectron. 58:242–248. 2014. View Article : Google Scholar : PubMed/NCBI

17 

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. Oncol Rep. 31:2377–2384. 2014.PubMed/NCBI

18 

Fujii Y, Kimura M, Ishii Y, Yamamoto R, Morita R, Hayashi SM, Suzuki K and Shibutani M: Effect of enzymatically modified isoquercitrin on preneoplastic liver cell lesions induced by thioacetamide promotion in a two-stage hepatocarcinogenesis model using rats. Toxicology. 305:30–40. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Shimada Y, Dewa Y, Ichimura R, Suzuki T, Mizukami S, Hayashi SM, Shibutani M and Mitsumori K: Antioxidant enzymatically modified isoquercitrin suppresses the development of liver preneoplastic lesions in rats induced by β-naphthoflavone. Toxicology. 268:213–218. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Amado NG, Cerqueira DM, Menezes FS, da Silva JF, Neto VM and Abreu JG: Isoquercitrin isolated from Hyptis fasciculata reduces glioblastoma cell proliferation and changes β-catenin cellular localization. Anticancer Drugs. 20:543–552. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Ono H, Basson MD and Ito H: PTK6 promotes cancer migration and invasion in pancreatic cancer cells dependent on ERK signaling. PLoS One. 9:e960602014. View Article : Google Scholar : PubMed/NCBI

22 

Takahashi R, Hirata Y, Sakitani K, Nakata W, Kinoshita H, Hayakawa Y, Nakagawa H, Sakamoto K, Hikiba Y, Ijichi H, Moses HL, Maeda S and Koike K: Therapeutic effect of c-Jun N-terminal kinase inhibition on pancreatic cancer. Cancer. 104:337–344. 2013.

23 

Hornick JR, Vangveravong S, Spitzer D, Abate C, Berardi F, Goedegebuure P, Mach RH and Hawkins WG: Lysosomal membrane permeabilization is an early event in sigma-2 receptor ligand mediated cell death in pancreatic cancer. J Exp Clin Cancer Res. 31:412012. View Article : Google Scholar : PubMed/NCBI

24 

Zagon IS and McLaughlin PJ: Targeting opioidergic pathways as a novel biological treatment for advanced pancreatic cancer. Expert Rev Gastroenterol Hepatol. 6:133–135. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Oliveira FQ, Andrade-Neto V, Krettli AU and Brandão MG: New evidences of antimalarial activity of Bidens pilosa roots extract correlated with polyacetylene and flavonoids. J Ethnopharmacol. 93:39–42. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Atta AH and Mouneir SM: Evaluation of some medicinal plant extracts for antidiarrhoeal activity. Phytother Res. 19:481–485. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Sukumaran P, Nair AG, Chinmayee DM, Mini I and Sukumaran ST: Phytochemical investigation of Bidens biternata (Lour.) Merr and Sheriff - a nutrient-rich leafy vegetable from Western Ghats of India. Appl Biochem Biotechnol. 167:1795–1801. 2012. View Article : Google Scholar : PubMed/NCBI

28 

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. View Article : Google Scholar : PubMed/NCBI

29 

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

30 

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

31 

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. View Article : Google Scholar : PubMed/NCBI

32 

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. View Article : Google Scholar

33 

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). PubMed/NCBI

34 

Lee S, Park HS, Notsu Y, Ban HS, Kim YP, Ishihara K, Hirasawa N, Jung SH, Lee YS, Lim SS, Park EH, Shin KH, Seyama T, Hong J and Ohuchi K: Effects of hyperin, isoquercitrin and quercetin on lipopolysaccharide-induced nitrite production in rat peritoneal macrophages. Phytother Res. 22:1552–1556. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Hirano T, Kawai M, Arimitsu J, Ogawa M, Kuwahara Y, Hagihara K, Shima Y, Narazaki M, Ogata A, Koyanagi M, Kai T, Shimizu R, Moriwaki M, Suzuki Y, Ogino S, Kawase I and Tanaka T: Preventative effect of a flavonoid, enzymatically modified isoquercitrin on ocular symptoms of Japanese cedar pollinosis. Allergol Int. 58:373–382. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Magalingam KB, Radhakrishnan A and Haleagrahara N: Protective effects of flavonol isoquercitrin, against 6-hydroxy dopamine (6-OHDA)-induced toxicity in PC12 cells. BMC Res Notes. 7:492014. View Article : Google Scholar : PubMed/NCBI

37 

Li R, Yuan C, Dong C, Shuang S and Choi MM: In vivo antioxidative effect of isoquercitrin on cadmium-induced oxidative damage to mouse liver and kidney. Naunyn Schmiedebergs Arch Pharmacol. 383:437–445. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Gong WY, Wu JF, Liu BJ, Zhang HY, Cao YX, Sun J, Lv YB, Wu X and Dong JC: Flavonoid components in Scutellaria baicalensis inhibit nicotine-induced proliferation, metastasis and lung cancer-associated inflammation in vitro. Int J Oncol. 44:1561–1570. 2014.PubMed/NCBI

39 

Bądziul D, Jakubowicz-Gil J, Paduch R, Głowniak K and Gawron A: Combined treatment with quercetin and imperatorin as a potent strategy for killing HeLa and Hep-2 cells. Mol Cell Biochem. 392:213–227. 2014. View Article : Google Scholar :

40 

You OH and Kim SH, Kim B, Sohn EJ, Lee HJ, Shim BS, Yun M, Kwon BM and Kim SH: Ginkgetin induces apoptosis via activation of caspase and inhibition of survival genes in PC-3 prostate cancer cells. Bioorg Med Chem Lett. 23:2692–2695. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Mujumdar N, Banerjee S, Chen Z, Sangwan V, Chugh R, Dudeja V, Yamamoto M, Vickers SM and Saluja AK: Triptolide activates unfolded protein response leading to chronic ER stress in pancreatic cancer cells. Am J Physiol Gastrointest Liver Physiol. 306:G1011–G1020. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Heilmann AM, Perera RM, Ecker V, Nicolay BN, Bardeesy N, Benes CH and Dyson NJ: CDK4/6 and IGF1 receptor inhibitors synergize to suppress the growth of p16INK4A-deficient pancreatic cancers. Cancer Res. 74:3947–3958. 2014. View Article : Google Scholar : PubMed/NCBI

43 

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

44 

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

45 

Kim H, Lee SW, Park JS, Min JH and Kim HK: Genomic analysis of [d-Ala2, d-Leu5] enkephalin preconditioning in cortical neuron and glial cell injury after oxygen deprivation. Brain Res. 1447:91–105. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Galeotti N and Ghelardini C: Regionally selective activation and differential regulation of ERK, JNK and p38MAP kinase signalling pathway by protein kinase C in mood modulation. Int J Neuropsychopharmacol. 15:781–793. 2012. View Article : Google Scholar

47 

Raman M, Chen W and Cobb MH: Differential regulation and properties of MAPKs. Oncogene. 26:3100–3112. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Robbs BK, Lucena PI and Viola JP: The transcription factor NFAT1 induces apoptosis through cooperation with Ras/Raf/MEK/ERK pathway and upregulation of TNF-α expression. Biochim Biophys Acta. 1833:2016–2028. 2013. View Article : Google Scholar : PubMed/NCBI

49 

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

50 

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

51 

Alspach E, Flanagan KC, Luo X, Ruhland MK, Huang H, Pazolli E, Donlin MJ, Marsh T, Piwnica-Worms D, Monahan J, Novack DV, McAllister SS and Stewart SA: p38MAPK plays a crucial role in stromal-mediated tumorigenesis. Cancer Discov. 4:716–729. 2014. View Article : Google Scholar : PubMed/NCBI

52 

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

Related Articles

Journal Cover

February-2015
Volume 33 Issue 2

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
Chen Q, Li P, Xu Y, Li Y and Tang B: Isoquercitrin inhibits the progression of pancreatic cancer in vivo and in vitro by regulating opioid receptors and the mitogen-activated protein kinase signalling pathway Retraction in /10.3892/or.2023.8505. Oncol Rep 33: 840-848, 2015
APA
Chen, Q., Li, P., Xu, Y., Li, Y., & Tang, B. (2015). Isoquercitrin inhibits the progression of pancreatic cancer in vivo and in vitro by regulating opioid receptors and the mitogen-activated protein kinase signalling pathway Retraction in /10.3892/or.2023.8505. Oncology Reports, 33, 840-848. https://doi.org/10.3892/or.2014.3626
MLA
Chen, Q., Li, P., Xu, Y., Li, Y., Tang, B."Isoquercitrin inhibits the progression of pancreatic cancer in vivo and in vitro by regulating opioid receptors and the mitogen-activated protein kinase signalling pathway Retraction in /10.3892/or.2023.8505". Oncology Reports 33.2 (2015): 840-848.
Chicago
Chen, Q., Li, P., Xu, Y., Li, Y., Tang, B."Isoquercitrin inhibits the progression of pancreatic cancer in vivo and in vitro by regulating opioid receptors and the mitogen-activated protein kinase signalling pathway Retraction in /10.3892/or.2023.8505". Oncology Reports 33, no. 2 (2015): 840-848. https://doi.org/10.3892/or.2014.3626