Ursolic acid inhibits growth and induces apoptosis in gemcitabine-resistant human pancreatic cancer via the JNK and PI3K/Akt/NF-κB pathways

  • Authors:
    • Jingjie Li
    • Xiaoyan Liang
    • Xing Yang
  • View Affiliations

  • Published online on: May 22, 2012     https://doi.org/10.3892/or.2012.1827
  • Pages: 501-510
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pancreatic cancer is one of the most deadly carcinomas worldwide. Although gemcitabine as the standard chemotherapy agent has been proven to be effective, the response rate remains at 5.4% and the 5-year survival rate is extremely poor. Ursolic acid (UA) is a small molecule compound extracted from Chinese herbs as well as edible vegetables and a well-known anti-inflammatory and immunosuppressive agent. Here, we show that UA has potential to be developed into an anti-neoplastic agent against gemcitabine-resistant pancreatic cancer and to explore its molecular mechanism of action. In vitro, we used three different malignancy grades of pancreatic resistant cancer cell lines including MIA PaCa-2, PANC-1 and Capan-1 to assess the antitumor effect of UA. We found that UA inhibited growth and induced apoptosis in a dose-dependent manner in all of the three pancreatic cancer cell lines. Both extrinsic and intrinsic pathways were found to be involved in apoptotic cascade. The potential signaling pathways are concerned with inactivation of the PI3K/Akt/NF-κB pathway and activation of the c-Jun-terminal kinase (JNK) pathway. The JNK inhibitor SP600125 partly abrogated the caspase-9 activation caused by UA. The Akt inhibitor LY294002 did not mimic the effect of UA on caspase-8 and -9, but inhibited the viability of MIA PaCa-2 cells to some extent. Equally, UA also overcame the chemoresistance in the chemoresistant endometrial and ovarian carcinoma cell lines (HEC-1A and OVCAR-3). Moreover, UA caused cytotoxicity to a nude mouse xenograft model in vivo. Therefore, our present data suggest that UA can act as a novel and potent therapeutic agent in gemcitabine-resistant pancreatic cancer and even as a promising candidate in other chemoresistant cancers.

Introduction

Pancreatic cancer, characterized by aggressive malignancy and high resistance to chemotherapy agents, is considered as one of the most lethal tumors with a 5-year mortality of 97–98% (1,2). Surgery is the only curative treatment for pancreatic cancer. However, <20% patients have resectable tumor at the time of diagnosis due to the poor diagnosis (3). Except for the surgical therapy, both of chemotherapy and radiation regimens hardly ameliorate the outcome of pancreatic cancer patients, which causes ~90% of cases death within 1 year after diagnosis (4). Since the end of 20th century, gemcitabine, a deoxycitidine analogue, was confirmed to improve the objective response rate in patients with advanced pancreatic carcinoma, and has been applied as a first-line chemotherapy agent (5,6). However, only 5.4% of all patients are sensitive to the gemcitabine-based chemotherapy (7,8). Thus, novel chemotherapeutic drugs and candidate compounds are urgently needed to improve the outcome of patients with the lethal malignancy.

Ursolic acid (UA) is a pentacyclic triterpenoid compound extracted and purified from many types of medicinal plants, herbs, and edible vegetables (9). UA contains a variety of pharmacological activities. The anti-oxidative and anti-inflammatory actions of UA are widely applied to hepatoprotection, neuroprotection, cardiovascular protection and so on (1014). Besides these effects, UA has been proved to possess multiple anti-tumor activities including inhibiting tumorigenesis and progression (9). UA causes cytotoxicity to diverse malignancies such as fibrosarcoma, prostate cancer, hepatocellular carcinoma, colorectal cancer, melanoma, and breast cancer (1520). However, the cytotoxic role of UA in gemcitabine-resistant human pancreatic cancer and its potential molecular mechanisms are still unclear.

In this study, we investigated the effect of UA on growth restriction and apoptosis in human pancreatic resistant cancer in vitro and in vivo. Additionally, we explored possible mechanisms of action, such as PI3K/Akt/NF-κB and c-Jun N-terminal kinase (JNK) pathways.

Materials and methods

Cell culture and reagents

The human pancreatic resistant cancer cell lines MIA PaCa-2 and PANC-1 were kindly provided by Shanghai Institute of Materia Medica. The human pancreatic resistant cancer cell line Capan-1 was kindly provided by Department of Pharmacology, Sun Yat-Sen University. The paclitaxel-resistant endometrial cell line HEC-1A was purchased from Cell Bank of Chinese Academy of Sciences. The platinum-resistant ovarian cancer cell line OVCAR-3 was purchased from Experimental Animal Center of Sun Yat-Sen University. The cells were routinely cultured in DMEM (Invitrogen, Grand Island, NY) supplemented with 10% fetal bovine serum (FBS) (Invitrogen), 100 U/ml penicillin, and 100 mg/ml streptomycin in 5% CO2 at 37°C. Ursolic acid (UA) (National Institutes for Food and Drug Control, China) was dissolved in DMSO (Sigma-Aldrich, St. Louis, MO) as a 100 mM stock solution and stored at −20°C. Further dilution was done in serum-free cell culture medium. PI3K inhibitor LY294002 (Cell Signaling Technology, Beverly, MA) was dissolved in DMSO (Sigma) as a 50 mM stock solution and stored at −20°C. Similarly, JNK inhibitor SP600125 (Sigma) was dissolved and stored as a 10 mM stock solution by DMSO at −20°C.

Cell viability and proliferation

Cells were seeded into 96-well plates at a density of 2×103/well and allowed to attach overnight before being treated by different concentrations of UA for 24 h. Then cell viability and proliferation were evaluated. Cell viability was tested by the 3-(4,5-dimethylthiaziazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT, Sigma) assay as described in the literature (21). Detection of cell proliferation was through the colorimetric 5′-bromo-2′-deoxy-uridine (BrdU) incorporation assay (Roche Diagnostic Corp., Indianapolis, IN, USA) according to the instruction of the manufacturer. Briefly, cells were labeled by addition of BrdU for 4 h in 5% CO2 at 37°C. Then, FixDenat solution was used for cells fixation. The anti-BrdU-POD antibody was added to locate the BrdU label in the DNA. After the incubation of the peroxidase substrate, the immune complex was detected by a multi-well spectrophotometer reader using an iMark™ Microplate Reader (Bio-Rad, Richmond, CA).

Hoechst 33258 staining

The three types of cells at the density of 2×106/well in a 6-well plate were fixed by 4% paraform for 10 min. Then Hoechst 33258 (Sigma) at 5 μg/ml was added to the cells for 15 min at room temperature in the dark. The nuclei morphology was observed by fluorescence microscopy (Olympus, Melvie, NY) with a 340-nm excitation filter.

Caspase-3/7, −8 and −9 activity measurements

Caspase-3/7, −8 and −9 activity were assessed by Caspase-Glo luminescent-based assays (Promega, Madison, WI). Accordance with the instructions, cells (1×104/well) were seeded into a 96-well plate overnight to ensure adherence. After various concentrations of UA treatment for 24 h in a 5% CO2-humidified atmosphere at 37°C, 100 μl of 3/7, −8 and −9 Caspase-Glo reagents were added to each well, respectively. Mixed and incubated for 1 h, the mixtures were transferred to a fluorescence microtiter plate and quantified fluorescence intensity by a luminometer. The corresponding OD values from MTT assay described above were considered as the viable cell number to normalize the fluorescence intensity.

Cytochrome C ELISA assay

Cytoplasmic cytochrome C levels were determined by a cytochrome C ELISA kit (R&D Systems, Minneapolis, MN). The cytoplasmic protein was collected by a nuclear and cytoplasmic extraction reagents (Pierce, Rockford, IL) from adherent cells (2×106/well) in a 6-well plate after UA incubation for 24 h. The BCA protein assay kit (Pierce) was used to normalize the cytoplasmic protein. The protein was stored in −80°C until it was analyzed by cytochrome C ELISA kit according to the manufacturer's instructions.

Western blotting and immunofluorescent analysis

Western blotting was performed as described previously (22). The following antibodies were used: antibodies against PI3K, Akt, phospho-Akt, GSK-3β, phospho-GSK-3β, NF-κB phospho-p65, JNK, phospho-SAPK/JNK, phospho-MDM2, IκB (1:1000, Cell Signaling Technology); NF-κB p65, phospho-IκB (1:1000, Santa Cruz Biotechnology, Santa Cruz, CA), and actin (1:1000, Thermo Scientific IHC, Fremont, CA).

The immunofluorescent analysis was performed as previous protocol (23). After UA treatments, the MIA PaCa-2 cells were fixed in 4% paraformaldehyde for 30 min and perforated by PBS containing 0.1% Triton X-100. The cells were blocked with 5% bovine serum albumin (BSA) for 1 h at 37°C and incubated the NF-κB p65 antibody (1:200) at 4°C overnight. After washing by PBS, Cy3-conjugated goat anti-rabbit IgG (1:50, Proteintech, Chicago, IL) was used as the secondary antibody for 1 h at 37°C. Nuclear staining was achieved by incubating the cells in DAPI for 10 min. The fluorescence was photographed with fluorescence microscopy (Olympus).

Mouse xenograft model and immunohistochemical analysis (IHC)

To establish a subcutaneous xenograft model, MIA PaCa-2 cells (5×106) were inoculated subcutaneously on the flanks of 6-week-old nude mice (female, purchased from Experimental Animal Center of Southern Medical University). When the average tumor size reached ~100 mm3, mice with tumor xenografts were randomized into three groups with six mice in each group including: i) vehicle alone (normal saline with 1% DMSO i.p.), ii) low dose UA (100 mg/kg twice a week i.p.), iii) high dose UA (200 mg/kg twice a week i.p.). All of the treatments were administered at a dose of 0.1 ml/10 g bodyweight. The formula, (length × width2)/2, was used to estimate tumor sizes. Both the body weight and tumor sizes were recorded twice a week. At the end of the experiment, the animals were sacrificed and the tumors were dissected and weighed to calculate the inhibitory rate. Animal studies were performed according to the institutional guidelines of Experimental Animal Center in Sun Yat-sen University.

The paraffin tissue sections were obtained from the tumor tissues from the nude mice, IHC were performed as described previously (24). Briefly, the sections were deparaffinizated with xylene and microwaved for 10 min in the presence of 10 mM citric acid buffer. After quenching the endogenous peroxidases by hydrogen peroxide, the sections were blocked by 5% BSA/0.3% Triton X-100 for 20 min. Then the sections were incubated with cleaved caspase-3 (1:200) antibody overnight at 4°C and secondary antibody, goat anti-rabbit IgG (1:50, Proteintech) for 30 min at 37°C, and then signal was generated by using the DAB substrate kit (Boster, Wuhan, China) for peroxidase.

Statistical analysis

Data are expressed as mean ± standard deviation (SD) of three separate experiments and analyzed by SPSS. P<0.05 was considered significant and determined by the two-sample Student's t-test or one-factor ANOVA analysis.

Results

Ursolic acid (UA) decreases the viability and proliferation of the human pancreatic resistant cancer cell lines

In order to appraise the cytotoxicity of UA on pancreatic-resistant carcinoma, three different malignancy grades of pancreatic resistant cancer cell lines including MIA PaCa-2 (grade 3), PANC-1 (grade 2) and Capan-1 (grade 1) were exposed to increasing concentrations of UA (15, 30, 45 and 60 μM) for 24 h (6,2527). Firstly, the morphology of three cell lines under microscopic observation revealed the corresponding reduction of surviving cell numbers and morphological changes after UA treatments (Fig. 1A). In addition, cell viability was analyzed by MTT assay. As shown in Fig. 1B, UA dose-dependently decreased the viability of three cell lines with the half maximal inhibitory concentration (IC50 value) at 40.8, 45.3 and 41.3 μM, respectively. Gemcitabine as the positive control at the concentration of 45 μM did not induce significant cytotoxicity to PANC-1 cells comparing with the vehicle for 24 h. However, UA decreased the viability by 62.5% at the same concentration in the identical cell lines. Similarly, UA achieved stronger cytotoxic effect than gemcitabine in MIA PaCa-2 and Capan-1 cells. The proliferation inhibition of UA was further investigated by 5′-bromo-2′-deoxy-uridine (BrdU) incorporation assay. As observed in Fig. 1C, growth inhibition effect of UA in MIA PaCa-2 cells started at 15 μM and increased up to 60 μM. The results were in agreement with the outcomes of MTT assay. These data suggested that UA was capable of causing growth inhibition to pancreatic-resistant carcinoma in vitro.

Both intrinsic and extrinsic pathways are involved in UA-triggered apoptosis

UA has been shown to induce apoptosis in multiple cancer cell lines (1620). To identify whether UA has the same effect in pancreatic-resistant cancer cell lines, the morphologic analysis was performed by Hoechst 33258 staining. MIA PaCa-2, PANC-1 and Capan-1 cells were exposed to various concentrations (15, 30, 45 and 60 μM) of UA for 24 h, the morphological characteristics of apoptotic cells such as condensed nuclei and cell shrinkage were discovered and apoptotic proportion grew with upward dose (Fig. 2A). To confirm the UA-triggered apoptosis in three cell lines, another apoptotic marker, caspase-3/7 activity, was examined. Clearly, the caspase-3/7 activity was promoted gradually along with the rising concentrations of UA. Fig. 2B shows that the caspase-3/7 activity was enhanced 20-, 16- and 8-fold, respectively, in MIA PaCa-2, PANC-1 and Capan-1 cells at 60 μM UA.

The caspase-dependent cascade is mediated by endogenous and exogenous signal transductions (22). To study by which UA induces caspase-3/7 activation, several markers belonging to the two pathways were inspected. Caspase-8 is a downstream apoptotic molecule of the death receptor, which mediates the death receptor signaling pathway (extrinsic pathway). As shown in Fig. 2C, caspase-8 activity increased by ~15-fold with 60 μM UA treatment. Cytochrome C released from mitochondria into cytoplasm was the initial step in the intrinsic apoptotic pathway. Fig. 2D exhibits the cytochrome C relative release rate increase by ~4.5-fold at 60 μM UA. Besides, caspase-9 activity, another marker of the intrinsic apoptotic pathway, was also increased by ~14-fold after 60 μM UA intervention (Fig. 2E).

c-Jun-terminal kinase (JNK) pathway is potentially involved in UA-induced endogenous apoptosis

The mitogen-activated protein kinase (MAPK) family is closely related to cell apoptosis (28). There are some reports revealing that UA is able to activate JNK, which is one of crucial components in MAPK signaling pathway (16,29). Here, we detected JNK and P38 expression levels, both of which were reported as vital molecules of MAPK family in the programmed cell death, to explore more potential signal channels (30). We found that JNK activity, measured by phosphorylated-JNK level, was significantly up-regulated at the presence of 30 and 45 μM UA. But, UA had no obvious effect on JNK, P38 and phosphorylated-P38 protein levels (Fig. 3A). This result indicated that JNK may contribute to UA-induced apoptosis. To further study the apoptotic pathway in which JNK was involved, we used the JNK inhibitor SP600125 to block the activity of JNK pathway. MIA PaCa-2 cells were pretreated with 10 μM SP600125 for 1 h, and then treated with 45 μM UA for 12 h. As shown in Fig. 3B and C, pretreatment by SP600125 partially inhibited the caspase-9 activity by 16%, while caspase-8 activity was not altered significantly. These data indicated that JNK is at least partly involved in UA-induced endogenous apoptotic signaling transduction.

UA inhibits the PI3K/Akt pathway and down-regulates the expression of downstream proteins

To ascertain the potential mechanism by which UA caused growth suppression and apoptosis to pancreatic cancer, we also examined the activity of PI3K/Akt pathway in MIA PaCa-2 cell line. When cells were in the presence of increasing concentrations of UA (15, 30 and 45 μM) for 12 h, the results by western blotting demonstrated the total PI3K and the phosphorylated Akt levels were attenuated gradually in a dose-dependent manner, while the total Akt protein expressions did not alter (Fig. 4A). PI3K/Akt pathway is constitutively activated in most tumor cell lines and regulates a series of biological characteristics of tumors such as growth and apoptosis (31). Therefore, we checked the possible Akt targeting effector molecules downstream. Similar changes to Akt, were recorded for the phosphorylation level of GSK-3β, MDM2, P65, IκB that also decreased gradually, whereas the total proteins of GSK-3β, P65, IκB did not show any corresponding alteration (Fig. 4B).

PI3K/Akt/NF-κB pathway is involved in UA-induced cell viability decrease, but not intrinsic and extrinsic apoptosis

Aberrant activation of NF-κB greatly contributes to chemoresistance in pancreatic cancer, and suppression of NF-κB sensitizes the treatment outcome of gemcitabine (32). To assess whether the UA-induced NF-κB activation is a downstream event of PI3K/Akt signaling pathway, IκB and its phosphorylated form were further investigated. We inhibited the phosphorylation of Akt by 10, 20, 40 μM LY294002 for 12 h. As seen in Fig. 4C, 20 μM LY294002 and above abolished more than half of the phosphorylated level of Akt protein. Phosphorylated IκB was evidently modulated downward in a dose-dependent pattern. However, there was no alteration of total IκB within 12 h. In addition, the nuclear translocation of the cytoplasmic NF-κB p65 subunit was also checked by immunofluorescence. UA (45 μM) and/or 40 μM LY294002 for 12 h repressed the NF-κB p65 nuclear translocation compared with vehicle (Fig. 4D). These results suggested there was crosstalk between PI3K/Akt and NF-κB signaling pathways in MIA PaCa-2 cells.

We next examined the involvement of PI3K/Akt/NF-κB pathway in the apoptotic cascade. The MIA PaCa-2 cells were incubated with 40 μM LY294002 for 12 h. As seen in Fig. 4E and F, inhibiting the activation of Akt can not activate caspase-8 and −9. Hence, PI3K/Akt/NF-κB pathway is not the main participant in UA-induced intrinsic and extrinsic apoptotic signaling transductions. However, the cell viability was decreased by ~25% after exposure to 40 μM LY294002 for 24 h. These findings indicated that PI3K/Akt/NF-κB pathway was involved in UA-induced cytotoxicity to MIA PaCa-2 cells, but the mechanism was not caspase-8- and −9-dependent.

UA overcomes chemoresistance in other resistant cancer cell lines

We also tested whether UA possessed the potential to induce other resistant cancer cells death in the similar fashion. A paclitaxel-resistant endometrial cell line (HEC-1A) and a platinum-resistant ovarian cancer cell line (OVCAR-3) were used to examine the cell viability after UA exposure (33,34). As seen in Fig. 5A and B, UA inhibited the cell viability in a time and dose-dependent manner. Moreover, UA also increased the phosphorylated JNK, and decreased the expression of phosphorylated Akt (Fig. 5C and D). These results further confirmed that the mechanism of action of UA was associated with JNK and Akt pathways.

UA causes tumor regression in MIA PaCa-2 xenograft model

The data described above clearly showed that UA was a potent compound to inhibit proliferation and caused deaths of various chemotherapy-resistant cancer cell lines. Thus we tried to apply UA in a nude mouse xenograft model to provide evidence for clinical application. Until the tumor volume reached ~100 mm3, the mice were treated i.p. with either vehicle or UA at 100/200 mg/kg twice a week. Significant inhibition of tumor growth by UA was observed after UA treatment for 20 days (Fig. 6A and B). Tumor weights in UA-treated groups were also lower than the vehicle group (30% less in 100 mg/kg group and 50% less in 200 mg/kg group) (Fig. 6C). In order to measure the safety of UA, body weights of mice were regularly detected. As seen in Fig. 6D, there were no significant differences compared with control group after the UA treatment. Furthermore, we checked the cleavage caspase-3 expression by immunohistochemistry to reveal that the tumor in UA-exposed group had more visible apoptosis. These data elucidated that UA caused resistant pancreatic cancer death also in vivo.

Discussion

Due to the poor early diagnosis and serious chemotherapy resistance, pancreatic carcinoma is highly fatal. Although gemcitabine is known as the most active chemotherapy agent for this tumor, it has only a slight survival benefit and shows an objective tumor response rate of <10% owing to pre-existing or acquired chemo-resistance (35,36), thus demanding urgent development of effective therapeutic options. Here we demonstrate that a small molecule ursolic acid (UA), a well known anti-oxidative and anti-inflammatory agent, significantly suppresses the growth and induces apoptosis of resistant pancreatic cancer through activation of c-Jun-terminal kinase (JNK) pathway and inhibition of PI3K/Akt/NF-κB pathway.

UA, belonging to pentacyclic triterpenoid, is a soluble, natural, and small molecule compound, which has displayed its proliferation-inhibiting and apoptosis-inducing effects in some chemotherapy-resistant tumors (16,17,37). Previous study showed that UA only decreased the cell viability by ~40% in a hormone refractory prostate cancer cell line at 72 h (16). In contrast to the prostate cancer cells, UA exhibits much more potent cytotoxicity to pancreatic tumors. The half maximal inhibitory concentration (IC50 value) of UA was 40.8 μM in highly malignant pancreatic cancer cells (MIA PaCa-2 cells) within 24 h in vitro. In our data, both modulation of proliferation and induction of apoptosis by UA were observed within the time course of 24 h. However, UA had suppressed the proliferation significantly at 15 μM rather than any type of caspase activity or cytochrome C relative release rate. The alterations of proliferation inhibiting rate and caspase-3/7 activity were detected at 30–45 μM UA. Furthermore, UA at the higher concentration (60 μM) was only perceived to be effective in apoptosis induction. Hence, the results seem to indicate that decreased proliferation and increased apoptosis by UA together impact cell viability of pancreatic cancer cells.

The JNK pathway reported as important apoptosis inducing factor exerts antagonistic effects on apoptosis (30). JNK, a mitogen-activated protein kinase (MAPK), can be activated primarily by exposure to environmental stress (38). Although the extracellular signal-regulated kinase (ERK) pathway is considered as a great contributor to oncogenesis, previous study has demonstrated that it plays a lesser role in mitogen-induced survival of pancreatic cancer (39). However, many studies have reported that JNK pathway plays a crucial role in UA-induced apoptosis in many cancers (16,29,40). In our study, the activation of JNK, but not P38, was induced by UA in a dose-dependent manner in MIA PaCa-2 cells. Two major apoptotic signaling transductions, namely the receptor-mediated/exogenous and mitochondrial/endogenous pathways, are activated in UA-induced apoptosis in MIA PaCa-2 cells. The trigger of endogenous pathway depends on the release of cytochrome C from mitochondria to cytosol and activation of caspase-9 (41). In the extrinsic pathway, the binding of death ligands to their homologous receptors drives activity of caspase-8 (42). Here, we showed that SP600125, a specific JNK inhibitor, could partly protect UA-induced caspase-9 activation rather than caspase-8. Our data are consistent with a previous report (16). Teraishi et al(43) have proved that JNK activation is required for gemcitabine to achieve its cytotoxicity and the weaker activity of JNK may result in gemcitabine resistance. Our data displayed that JNK pathway was activated excessively in the gemcitabine resistant cell line (MIA PaCa-2). We proved similarly that JNK pathway participated in UA-induced mitochondrial apoptotic signaling transduction in MIA PaCa-2 cells. Thus we speculate that UA-induced JNK activation may contribute to overcome gemcitabine resistance and induces programmed cell death in gemcitabine resistant pancreatic cancer.

The current consensus to PI3K/Akt signaling transduction is that this cascade contributes greatly to tumor progression and chemotherapy resistance (44). PI3K is a broadly expressed lipid kinase, the main action of which is to execute the catalyzed reaction to synthesize the second messenger-PIP3 from PIP2(44,45). Akt, acting downstream of PI3K, is usually activated by directly binding to PIP3 through its pleckstrin-homology (PH) domain and phosphorylation (44). Akt activation is closely relevant to anti-apoptosis and proliferative signal transduction. Primarily, Akt stimulates the degradation of IκB which results in the nuclear translocation of P65 and activation of target genes (46). Akt also catalyzes phosphorylation of MDM to improve its function to bind to P53 and enhance p53 degradation (47,48). In the aspect of cell proliferative promotion, Akt inhibits the kinase activity of GSK3β by maintaining the phosphorylated level to protect cyclin D from degradation (49). Our results showed that UA strongly suppressed the activation of Akt and blocked the activity of its downstream molecules mentioned above.

We proved that suppressing the Akt activity by LY294002, a selective Akt inhibitor, down-regulated the NF-κB activation by reducing the p65 nuclear translocation and phosphorylating IκB in MIA PaCa-2 cells. We propose that PI3K/Akt pathway seems crosstalk with NF-κB pathway in pancreatic cancer cells. Arlt et al(32) has reported previously the evident correlation between basal NF-κB activity and gemcitabine resistance in pancreatic cancer cells. There is some evidence showing that PI3K/Akt inhibition promoted anti-tumor effectiveness of gemcitabine via inhibiting PI3K/Akt phosphorylation in vitro and in vivo(50,51). UA may demonstrate its potent anti-cancer effects via PI3K/Akt/NF-κB pathway in MIA PaCa-2 cells. We observed that LY294002 did not protect MIA PaCa-2 cells from caspase-8/9 activations. Therefore, exogenous and endogenous apoptosis are not affected equally by UA-triggered Akt activation. However, LY294002 decreased the viability of MIA PaCa-2 cells to some extent, and it is well-known that cell viability is a combination of cell proliferation and death. We argue that the UA-induced PI3K/Akt/NF-κB pathway suppression may only affect the pancreatic cancer cell proliferative rate. Besides, recent study also demonstrated that UA was able to induce tumor cells apoptosis through a caspase-independent pathway (17). That phenomenon means UA has the chance to apply for combined treatment to overcome chemotherapy resistance. In our study, we can not exclude that the PI3K/Akt/NF-κB pathway also triggers caspase-independent apoptosis. But further studies are needed to prove the hypothesis.

To examine whether the effect of UA on JNK and Akt pathway is confined to resistant pancreatic cancer cells solely or is a general phenomenon which is applicable to other resistant cancer cells, we examined the effect of UA on an endometrial cell line (HEC-1A) and an ovarian cancer cell line (OVCAR-3) because both of them are highly resistant to chemotherapy. According to our data, the JNK activity was inhibited, but Akt was excessively activated in these cells. UA significantly decreased the cell viability of HEC-1A cells and OVCAR-3 cells, which was associated with an increase of JNK activation and a decrease of Akt activation. For future clinical application, the safety and efficacy of UA were further detected in vivo. We presented evidence that UA suppressed tumor growth without any obvious side effects. As a natural compound extracted from edible foods and herbs, UA is relatively low-toxic with LD50 value >637 mg/kg intraperitoneally in mice. Our studies provide further evidence that UA is a potent compound to apply to overcome chemoresistance in oncological therapy.

In conclusion, UA is a small, safe, potent and multifuntional molecule which is easily produced and relatively safe. UA has been applied in clinical trial in some areas to treat cancer (52). Consequently, UA is comparable or even superior to conventional anti-tumor drugs, encouraging further the use of this small molecular compound in gemcitabine resistant pancreatic cancer and other chemo-insensitive cancers.

Acknowledgements

This study was supported by National Natural Science Foundation of China (no. 30973202), Doctoral Fund of Ministry of Education of China (no. 20090171110059), Natural Science Foundation of Guangdong Province (no. S2011010004621), and Guangdong Provincial Fund of Industry, Education and Academy (no. 2008B090500194).

References

1 

Stathis A and Moore MJ: Advanced pancreatic carcinoma: current treatment and future challenges. Nat Rev Clin Oncol. 7:163–172. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Campbell PJ, Yachida S, Mudie LJ, et al: The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature. 467:1109–1113. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Herrmann R and Jelic S: Pancreatic cancer: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 19(Suppl 2): I25–I26. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Rosewicz S and Wiedenmann B: Pancreatic carcinoma. Lancet. 349:485–489. 1997. View Article : Google Scholar : PubMed/NCBI

5 

Burris HR, Moore MJ, Andersen J, et al: Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 15:2403–2413. 1997.PubMed/NCBI

6 

Ou YQ, Zhu W, Li Y, et al: Aspirin inhibits proliferation of gemcitabine-resistant human pancreatic cancer cells and augments gemcitabine-induced cytotoxicity. Acta Pharmacol Sin. 31:73–80. 2010. View Article : Google Scholar

7 

Ina S, Hirono S, Noda T and Yamaue H: Identifying molecular markers for chemosensitivity to gemcitabine in pancreatic cancer: increased expression of interferon-stimulated gene 15 kDa is associated with intrinsic chemoresistance. Pancreas. 39:473–485. 2010. View Article : Google Scholar

8 

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

9 

Liu J: Oleanolic acid and ursolic acid: research perspectives. J Ethnopharmacol. 100:92–94. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Liu J: Pharmacology of oleanolic acid and ursolic acid. J Ethnopharmacol. 49:57–68. 1995. View Article : Google Scholar : PubMed/NCBI

11 

Shih YH, Chein YC, Wang JY and Fu YS: Ursolic acid protects hippocampal neurons against kainate-induced excitotoxicity in rats. Neurosci Lett. 362:136–140. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Chung YK, Heo HJ, Kim EK, et al: Inhibitory effect of ursolic acid purified from Origanum majorana L on the acetylcholinesterase. Mol Cells. 11:137–143. 2001.PubMed/NCBI

13 

Tsai SJ and Yin MC: Antioxidative and anti-inflammatory protection of oleanolic acid and ursolic acid in PC12 cells. J Food Sci. 73:H174–H178. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Somova LO, Nadar A, Rammanan P and Shode FO: Cardiovascular, antihyperlipidemic and antioxidant effects of oleanolic and ursolic acids in experimental hypertension. Phytomedicine. 10:115–121. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Cha HJ, Bae SK, Lee HY, et al: Anti-invasive activity of ursolic acid correlates with the reduced expression of matrix metalloproteinase-9 (MMP-9) in HT1080 human fibrosarcoma cells. Cancer Res. 56:2281–2284. 1996.PubMed/NCBI

16 

Zhang Y, Kong C, Zeng Y, et al: Ursolic acid induces PC-3 cell apoptosis via activation of JNK and inhibition of Akt pathways in vitro. Mol Carcinog. 49:374–385. 2010.PubMed/NCBI

17 

Yang L, Liu X, Lu Z, et al: Ursolic acid induces doxorubicin-resistant HepG2 cell death via the release of apoptosis-inducing factor. Cancer Lett. 298:128–138. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Xavier CP, Lima CF, Preto A, Seruca R, Fernandes-Ferreira M and Pereira-Wilson C: Luteolin, quercetin and ursolic acid are potent inhibitors of proliferation and inducers of apoptosis in both KRAS and BRAF mutated human colorectal cancer cells. Cancer Lett. 281:162–170. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Manu KA and Kuttan G: Ursolic acid induces apoptosis by activating p53 and caspase-3 gene expressions and suppressing NF-kappaB mediated activation of bcl-2 in B16F-10 melanoma cells. Int Immunopharmacol. 8:974–981. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Wang JS, Ren TN and Xi T: Ursolic acid induces apoptosis by suppressing the expression of FoxM1 in MCF-7 human breast cancer cells. Med Oncol. 29:10–15. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Zhu W, Ou Y, Li Y, et al: A small-molecule triptolide suppresses angiogenesis and invasion of human anaplastic thyroid carcinoma cells via down-regulation of the nuclear factor-kappa B pathway. Mol Pharmacol. 75:812–819. 2009. View Article : Google Scholar

22 

Li J, Zhu W, Leng T, et al: Triptolide-induced cell cycle arrest and apoptosis in human renal cell carcinoma cells. Oncol Rep. 25:979–987. 2011.PubMed/NCBI

23 

He D, Xu Q, Yan M, et al: The NF-kappaB inhibitor, celastrol, could enhance the anti-cancer effect of gambogic acid on oral squamous cell carcinoma. BMC Cancer. 9:3432009. View Article : Google Scholar : PubMed/NCBI

24 

Stairs DB, Nakagawa H, Klein-Szanto A, et al: Cdx1 and c-Myc foster the initiation of transdifferentiation of the normal esophageal squamous epithelium toward Barrett's esophagus. PLoS One. 3:e35342008. View Article : Google Scholar : PubMed/NCBI

25 

Xia Y, Liu Y, Wan J, et al: Novel triazole ribonucleoside down-regulates heat shock protein 27 and induces potent anticancer activity on drug-resistant pancreatic cancer. J Med Chem. 52:6083–6096. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Jonckheere N, Fauquette V, Stechly L, et al: Tumour growth and resistance to gemcitabine of pancreatic cancer cells are decreased by AP-2alpha overexpression. Br J Cancer. 101:637–644. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Miyamoto H, Murakami T, Tsuchida K, Sugino H, Miyake H and Tashiro S: Tumor-stroma interaction of human pancreatic cancer: acquired resistance to anticancer drugs and proliferation regulation is dependent on extracellular matrix proteins. Pancreas. 28:38–44. 2004. View Article : Google Scholar

28 

Chang L and Karin M: Mammalian MAP kinase signalling cascades. Nature. 410:37–40. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Zhang YX, Kong CZ, Wang LH, et al: Ursolic acid overcomes Bcl-2-mediated resistance to apoptosis in prostate cancer cells involving activation of JNK-induced Bcl-2 phosphorylation and degradation. J Cell Biochem. 109:764–773. 2010.PubMed/NCBI

30 

Wagner EF and Nebreda AR: Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 9:537–549. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Falasca M: PI3K/Akt signalling pathway specific inhibitors: a novel strategy to sensitize cancer cells to anti-cancer drugs. Curr Pharm Des. 16:1410–1416. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Arlt A, Gehrz A, Muerkoster S, et al: Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene. 22:3243–3251. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Wang X, Yang Y, Xu C, et al: CHFR suppression by hypermethylation sensitizes endometrial cancer cells to paclitaxel. Int J Gynecol Cancer. 21:996–1003. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Lange TS, Kim KK, Singh RK, Strongin RM, McCourt CK and Brard L: Iron(III)-salophene: an organometallic compound with selective cytotoxic and anti-proliferative properties in platinum-resistant ovarian cancer cells. PLoS One. 3:e23032008. View Article : Google Scholar : PubMed/NCBI

35 

Welch SA and Moore MJ: Combination chemotherapy in advanced pancreatic cancer: time to raise the white flag? J Clin Oncol. 25:2159–2161. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Mamaghani S, Patel S and Hedley DW: Glycogen synthase kinase-3 inhibition disrupts nuclear factor-kappaB activity in pancreatic cancer, but fails to sensitize to gemcitabine chemotherapy. BMC Cancer. 9:1322009. View Article : Google Scholar

37 

Shan JZ, Xuan YY, Ruan SQ and Sun M: Proliferation-inhibiting and apoptosis-inducing effects of ursolic acid and oleanolic acid on multi-drug resistance cancer cells in vitro. Chin J Integr Med. 17:607–611. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Weston CR and Davis RJ: The JNK signal transduction pathway. Curr Opin Cell Biol. 19:142–149. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Perugini RA, McDade TP, Vittimberga FJ and Callery MP: Pancreatic cancer cell proliferation is phosphatidylinositol 3-kinase dependent. J Surg Res. 90:39–44. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Zhang YX, Kong CZ, Wang HQ, Wang LH, Xu CL and Sun YH: Phosphorylation of Bcl-2 and activation of caspase-3 via the c-Jun N-terminal kinase pathway in ursolic acid-induced DU145 cells apoptosis. Biochimie. 91:1173–1179. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Kroemer G and Reed JC: Mitochondrial control of cell death. Nat Med. 6:513–519. 2000. View Article : Google Scholar

42 

Ashkenazi A and Dixit VM: Death receptors: signaling and modulation. Science. 281:1305–1308. 1998. View Article : Google Scholar : PubMed/NCBI

43 

Teraishi F, Zhang L, Guo W, et al: Activation of c-Jun NH2-terminal kinase is required for gemcitabine's cytotoxic effect in human lung cancer H1299 cells. FEBS Lett. 579:6681–6687. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Vivanco I and Sawyers CL: The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat Rev Cancer. 2:489–501. 2002. View Article : Google Scholar : PubMed/NCBI

45 

Garcia-Echeverria C and Sellers WR: Drug discovery approaches targeting the PI3K/Akt pathway in cancer. Oncogene. 27:5511–5526. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Kane LP, Shapiro VS, Stokoe D and Weiss A: Induction of NF-kappaB by the Akt/PKB kinase. Curr Biol. 9:601–604. 1999. View Article : Google Scholar : PubMed/NCBI

47 

Mayo LD and Donner DB: A phosphatidylinositol 3-kinase/Akt pathway promotes translocation of Mdm2 from the cytoplasm to the nucleus. Proc Natl Acad Sci USA. 98:11598–11603. 2001. View Article : Google Scholar : PubMed/NCBI

48 

Zhou BP, Liao Y, Xia W, Zou Y, Spohn B and Hung MC: HER-2/neu induces p53 ubiquitination via Akt-mediated MDM2 phosphorylation. Nat Cell Biol. 3:973–982. 2001. View Article : Google Scholar : PubMed/NCBI

49 

Diehl JA, Cheng M, Roussel MF and Sherr CJ: Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. Genes Dev. 12:3499–3511. 1998. View Article : Google Scholar : PubMed/NCBI

50 

Pham NA, Tsao MS, Cao P and Hedley DW: Dissociation of gemcitabine sensitivity and protein kinase B signaling in pancreatic ductal adenocarcinoma models. Pancreas. 35:E16–E26. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Ng SS, Tsao MS, Nicklee T and Hedley DW: Wortmannin inhibits pkb/akt phosphorylation and promotes gemcitabine antitumor activity in orthotopic human pancreatic cancer xenografts in immunodeficient mice. Clin Cancer Res. 7:3269–3275. 2001.PubMed/NCBI

52 

Sultana N: Clinically useful anticancer, antitumor, and antiwrinkle agent, ursolic acid and related derivatives as medicinally important natural product. J Enzyme Inhib Med Chem. 26:616–642. 2011. View Article : Google Scholar

Related Articles

Journal Cover

August 2012
Volume 28 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
Li J, Liang X and Yang X: Ursolic acid inhibits growth and induces apoptosis in gemcitabine-resistant human pancreatic cancer via the JNK and PI3K/Akt/NF-κB pathways. Oncol Rep 28: 501-510, 2012
APA
Li, J., Liang, X., & Yang, X. (2012). Ursolic acid inhibits growth and induces apoptosis in gemcitabine-resistant human pancreatic cancer via the JNK and PI3K/Akt/NF-κB pathways. Oncology Reports, 28, 501-510. https://doi.org/10.3892/or.2012.1827
MLA
Li, J., Liang, X., Yang, X."Ursolic acid inhibits growth and induces apoptosis in gemcitabine-resistant human pancreatic cancer via the JNK and PI3K/Akt/NF-κB pathways". Oncology Reports 28.2 (2012): 501-510.
Chicago
Li, J., Liang, X., Yang, X."Ursolic acid inhibits growth and induces apoptosis in gemcitabine-resistant human pancreatic cancer via the JNK and PI3K/Akt/NF-κB pathways". Oncology Reports 28, no. 2 (2012): 501-510. https://doi.org/10.3892/or.2012.1827