Skip to main content

Pachymic acid induces apoptosis via activating ROS-dependent JNK and ER stress pathways in lung cancer cells

Abstract

Background

Pachymic acid (PA), a lanostane-type triterpenoid from Poria cocos, has been reported to possess anti-emetic, anti-inflammatory, and anti-cancer properties. Nonetheless, the anti-tumor effect of PA in lung cancer cells remains unclear. Herein, we report the chemotherapeutic effects and underlying mechanisms of PA against human lung cancer.

Methods

The anti-proliferative ability of PA on lung cancer cells was assessed by MTT, colony formation and EdU proliferation assays. Flow cytometric analysis was used to detect cell cycle changes. Apoptosis was determined by annexin V/PI double-staining and the DNA ladder formation assays. The expressions of the apoptosis-related proteins were analysed by western blot. The in vivo efficacy of PA was measured using a NCI-H23 xenograft model in nude mice.

Results

PA exhibited anti-tumor effects in vitro accompanied by induction of G2/M phase arrest and apoptosis in NCI-H23 and NCI-H460 lung cancer cells. Mechanistically, our data showed that PA induced reactive oxygen species (ROS) production, resulting in the activation of both c-Jun N-terminal kinase (JNK) and endoplasmic reticulum (ER) stress apoptotic pathways in lung cancer cells. Moreover, blockage of ROS production reversed PA-induced JNK and ER stress activation. Finally, PA inhibited the growth of NCI-H23 xenograft tumors without causing any host toxicity, and inhibited cell proliferation and induction of apoptosis of tumor cells in tumor xenograft tissues.

Conclusions

In summary, our study demonstrates that PA induces apoptosis through activation of the JNK and ER stress pathways in human lung cancer cells. Our findings provide a rationale for the potential application of PA in lung cancer therapy.

Background

Lung cancer is a leading cause of cancer-related deaths worldwide [1], and it can be divided into two histological groups: non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). NSCLC accounts for almost 80% of lung cancer cases. Although many factors, including tobacco smoke, ionizing radiation and viral infection are known to increase the risk of NSCLC, the mechanisms involved in NSCLC formation remain largely unknown [2]. Despite improvements in tumor response to chemotherapy, the long-term survival rate for advanced lung cancer patients remains quite low [1], calling for a dire need of novel strategies to treat lung cancer with more active agents [3]. Herbal medicines have long been an important source for anti-cancer agents, and some of them are currently used in clinical practice [4]. In recent years, plant-derived compounds have been extensively screened to explore their potential anti-tumor activity. A variety of FDA approved anti-tumor agents, including docetaxel and topotecan, have demonstrated clinical utility based on ongoing investigations of natural products.

Exploring the precise molecular mechanisms involved in actions of anti-cancer agents has become an important approach for anti-cancer drug evaluation and development. Among these molecular mechanisms, apoptosis is a highly regulated process of cell death that serves to eliminate heavily damaged cells without injuring surrounding healthy cells, and its dysregulation underlies numerous pathological conditions including cancer [5]. Over the years, apoptosis has emerged as the major mechanism by which anti-cancer agents act to eliminate cancer cells [6]. Recent clinical data has revealed that restore deregulated apoptosis in cancer cells by anti-cancer drugs can significantly improve patient survival [7]. In this context, several plant compounds have been proven, in a more specific manner, to possess promising anti-cancer activity through their apoptosis-inducing effects [8, 9].

Pachymic acid (PA) is a lanostane-type triterpenoid from Poria cocos, also called Fuling, which is an important component for treatment of breast cancer metastasis in traditional Chinese medicine. Previous studies have shown that PA could induce apoptosis in prostate cancer cells [10]. Lots of evidences have revealed that certain triterpenoids have anti-cancer activity, especially lanostane-type triterpenoids [11, 12]. Another study has shown that PA could stimulate glucose uptake through enhance GLUT4 expression and translocation [13]. Despite the diverse studies conducted to investigate the biological activities of PA, the potential of PA against lung cancer is poorly defined. In this study, we demonstrated that PA induced G2/M phase arrest and cell apoptosis in lung cancer cells, via activating ROS-dependent JNK mitochondrial and ER stress pathways. PA also exhibited anti-tumor ability in vivo without causing any host toxicity. Our findings suggest that PA could be a promising agent for the treatment of lung cancer.

Results

Pachymic acid (PA) inhibits growth of human lung cancer cells in vitro

To explore the biological role of PA in lung cancer, we first performed a cell viability assay on NCI-H23 and NCI-H460 cells by MTT assay, treated with different concentrations of PA (0, 20, 40, 80 and 160 µM) for 24, 48 and 72 h. As shown in Fig. 1b, PA induced reduction of cell growth rate in a concentration- and time-dependent manner in both NCI-H23 and NCI-H460 cells. In addition, we further corroborated these effects by performing colony formation assays. After 24 h treatment with different concentrations of PA, NCI-H23 and NCI-H460 cells were cultured for another 10 days, and colonies counted. As shown in Fig. 1c, PA decreased colony formation in a concentration-dependent manner. Furthermore, EdU incorporation assays showed that the proliferation of NCI-H23 and NCI-H460 cells were significantly inhibited by PA relative to vehicle control (Fig. 1d). These observations indicated that PA inhibited growth of lung cancer cells in vitro.

Fig. 1
figure 1

Chemical structure of pachymic acid (PA) and its effects on cell growth of human lung cancer cells. a Chemical structure of PA. b NCI-H23 and NCI-H460 cells were treated with 0, 20, 40, 80 and 160 µM PA for 24, 48 or 72 h, cell viability was measured by MTT assay. Absorbance was read at 490 nm with averages from triplicate wells. c, d NCI-H23 and NCI-H460 cells were treated with 0, 20, 40 and 80 μM PA for 24 h, the effect of PA on the growth of lung cancer cells was examined by colony formation assay (c) and EdU incorporation assay (d). Data are presented as mean ± SD from three independent experiments. *P < 0.05, **P < 0.01 compared with the control group.

Pachymic acid (PA) induces G2/M cell cycle arrest and apoptosis in lung cancer cells

To examine whether the reduction in cell growth in NCI-H23 and NCI-H460 cells by PA treatment was related to the induction of cell cycle arrest, the lung cancer cells were treated with varying concentrations of PA for 24 h, followed by the cell cycle determination by flow cytometry. As shown in Fig. 2a, PA treatment caused a concentration-dependent increase of G2/M phase cell population in NCI-H23 and NCI-H460 lung cancer cells, indicating PA was able to arrest the cell cycle in G2/M phase. We further determined whether PA-induced cancer cell growth inhibition involved apoptosis. Flow cytometry using annexin V-PI staining analysis showed that treatment with different concentrations of PA for 24 h, the proportions of apoptotic cells were markedly increased in NCI-H23 and NCI-H460 cells (Fig. 2b). Furthermore, the effect of PA on cell apoptosis was tested by the DNA ladder formation assay. As shown in Fig. 2c, DNA laddering was observed after the NCI-H23 and NCI-H460 cells treated with PA for 24 h, suggesting that PA was able to induce the lung cancer cell apoptosis. Taken together, these data suggested that the inhibition of cell growth by PA associated with the induction of G2/M phase arrest and apoptosis.

Fig. 2
figure 2

PA induces cell cycle arrest and apoptosis in human lung cancer cells. a NCI-H23 and NCI-H460 cells were treated with PA (20, 40 or 80 µM) for 24 h, and the number of cells in G2/M phase was determined via flow cytometry. b, c NCI-H23 and NCI-H460 cells were treated with PA (20, 40 or 80 µM) for 24 h, and the cell apoptosis was determined by Annexin V-FITC/PI staining (b) and DNA ladder formation assay (c). Data are presented as mean ± SD from three independent experiments. **P < 0.01 compared with the control group.

ROS generation mediates PA-activated JNK-mitochondrial and ER stress apoptotic pathways

To further investigate the underlying mechanisms involved in PA-induced effects on apoptosis in lung cancer cells. NCI-H23 and NCI-H460 cells were used for the subsequent studies. The mitogen-activated protein kinase (MAPK) pathway has been extensively evaluated in lung cancer, from the perspective of biomarkers of outcome and targets of therapy [14]. Therefore, we first focused on MAPK pathway. Surprisingly, we found that PA treatment significantly activated all of three pathways of MAPKs, including JNK, ERK, and p38 (Fig. 3a). We then determined the roles of JNK, ERK, and p38 in PA-induced cytotoxicity using specific small-molecule inhibitors. NCI-H23 and NCI-H460 cells were pre-treated with JNK inhibitor SP600125, ERK inhibitor PD98059, or p38 inhibitor SB203580, respectively for 1 h, then exposed to PA for 24 h. Interestingly, PD98059 or SB203580 did not alter the cell viability, but JNK inhibitor SP600125 partially attenuated PA-induced cytotoxicity, indicating that JNK activation was associated with PA-induced cell death (Fig. 3b). JNK is a well-known regulator in mitochondrial apoptotic pathway [15]. We therefore examined the effects of PA on Bcl-2 family proteins by western blot analysis, the results showed that Bcl-2 level was decreased, whereas Bax level was increased in a concentration-dependent manner (Fig. 3a). These results suggested that PA-induced NCI-H23 and NCI-H460 cells apoptosis is at least partly mediated by JNK-mitochondrial pathway.

Fig. 3
figure 3

ROS generation mediates PA-activated apoptotic pathways. a NCI-H23 and NCI-H460 cells were treated with PA (20, 40 and 80 µM) for 24 h. The protein levels of p-JNK, p-ERK, p-p38, Bcl-2 and Bax were determined by western blot. b NCI-H23 and NCI-H460 cells were pre-treated with 20 µM JNK inhibitor (SP600125), p38 inhibitor (SB203580), ERK inhibitor (PD98059) for 1 h, or pre-treated with ROS inhibitor NAC (5 mM) for 2 h, then exposed to PA (80 µM) for 24 h. Cell viability was determined by MTT assay. c NCI-H23 and NCI-H460 cells were treated with PA (20, 40 and 80 µM) for 24 h. The protein levels of ATF4, ATF6, XBP-1 and CHOP were determined by western blot. d NCI-H23 and NCI-H460 cells were pre-treated with or without 5 mM NAC for 2 h before exposure to PA (80 μM) for 24 h. The protein levels of p-JNK, Bcl-2, Bax, ATF4, ATF6, XBP-1 and CHOP were determined by western blot. Data are presented as mean ± SD from three independent experiments. **P < 0.01 compared with the control group.

In addition to mitochondrial pathway, recent advances have highlighted the importance of the endoplasmic reticulum (ER) in cell death processes. Pharmacological interventions that effectively enhance tumor cell death through activating ER stress have attracted a great deal of attention for anti-cancer therapy [16]. Thus, we next examined the expressions of ER stress-related proteins, such as transcription factor 4 (ATF4), transcription factor 6 (ATF6), X-box binding proteins 1 (XBP-1), and C/EBP-homologous protein (CHOP) in PA-treated NCI-H23 and NCI-H460 cells. Of note, PA showed concentration-dependent activation of these four protein expressions (Fig. 3c), indicating that PA-induced cell apoptosis is also mediated by ER stress pathway.

Previous studies have reported that ROS generation could trigger cell apoptosis via activating both mitochondrial and ER stress pathways [17, 18]. Therefore, to identify the role of ROS in mediating anti-tumor effects of PA, an ROS inhibitor NAC was used, which is commonly used to inhibit ROS production and test ROS inducers, despite some recent studies mentioning that effects of NAC in other pathways [24, 25]. As shown in Fig. 3b, pre-treated with the NAC for 2 h, then exposed to PA for 24 h. NAC significantly attenuated NCI-H23 and NCI-H460 cells death induced by PA. These results indicated that ROS production mediated PA-induced cell death. Further, we tested the effect of the ROS inhibitor NAC on PA-induced activation of JNK and ER stress in NCI-H23 and NCI-H460 cells by western blot. As shown in Fig. 3d, NAC pre-treatment blocked the expression of p-JNK, Bax, ATF4, ATF6, XBP-1 and CHOP, and attenuated the decrease of Bcl-2 in PA-treated NCI-H23 and NCI-H460 cells. These results suggested that ROS generation mediated PA-activated apoptotic pathways and was a critical upstream regulator in anti-tumor activity of PA.

Anti-tumor activity of pachymic acid (PA) in vivo

To evaluate the anti-tumor activity of PA in vivo, we used human lung cancer NCI-H23 tumor xenograft models. As seen in Fig. 4a, PA significantly suppressed tumor growth at doses of 30 and 60 mg/kg for 21 days compared with the control group. In addition, there is no significant difference in body weight change among the vehicle group and PA-treated groups (Fig. 4b). Moreover, there were no significant differences in the histological findings among the treatment and control groups in any of the other tissues examined (lung, liver, kidney and spleen) (Fig. 4c). These data showed that PA exhibited potent anti-tumor activity and high safety in vivo.

Fig. 4
figure 4

PA inhibits tumor xenograft growth in vivo. a Anti-tumor activity of PA in nude mice bearing NCI-H23 tumors. Vehicle control and PA (10, 30 and 60 mg/kg) were administered for 3 weeks (5 days/week) by ip injection. Tumor volume was monitored. b Body weight of the mice. c H&E staining of the paraffin-embedded sections of various tissues (lung, liver, kidney and spleen) obtained from mice were performed to assess whether there were any abnormalities caused by the treatment. Scale bars are 100 μm. Data in the graphs represent the mean ± SD (n = 6). **p < 0.01 versus vehicle control.

Pachymic acid (PA) inhibits proliferation and induces apoptosis via activating JNK and ER stress pathways in vivo

To elucidate the mechanisms of PA-meditated anti-tumor efficacy in vivo, we conducted IHC analysis in the NCI-H23 tumor xenograft model. Uncontrolled tumor cell proliferation is a characteristic feature of most cancers. We therefore analysed the tumor xenografts for the potential anti-proliferative effects of PA, using immunohistochemical detection of cell proliferation marker Ki-67-positive cells. As shown in Fig. 5b, PA decreased the expression of Ki-67. In addition, to determine whether inhibition of tumor growth by administration PA is caused by the apoptosis of tumor cells in xenograft tissues, the apoptotic effect of PA on tumor tissues was identified by expression of the DNA fragment using TUNEL assay. The results showed that there were greater numbers of TUNEL-positive cells in the PA-treated samples, as compared with the numbers in the non-PA-treated samples (Fig. 5c). Furthermore, we determined the effects of PA on JNK and ER stress pathways in tumor xenograft samples. Immunohistochemical analysis suggested that administration of PA resulted in increased p-JNK and CHOP in the tumor xenografts (Fig. 5d, e). Taken together, consistent with our in vitro data, the above results showed that PA blocked proliferation and induced apoptosis via activating JNK and ER stress pathways in human lung tumor xenograft model.

Fig. 5
figure 5

Mechanisms of anti-tumor activity of PA in vivo. a–e PA was administered for 3 weeks by ip injection to nude mice bearing NCI-H23 xenograft tumors. At the end of the experiments, tumors were excised and processed for H&E staining (a), immunostaining and quantification for Ki-67 (b), TUNEL (c), p-JNK (d) and CHOP (e). Scale bars are 100 μm.

Discussion

Lung cancer is the leading cause of cancer-related deaths worldwide. Chemotherapy is one of the major treatment methods. However, the overall survival rate for advanced lung cancer patients is still low [1]. Therefore, the search for new anti-tumor agents that are more effective but less toxic has become a matter of great interest. Natural products have played an important role as an effective source of anti-tumor agents. It is estimated that up to 30–40% of the anti-cancer drugs used globally are derived from plant sources [19]. Pachymic acid (PA), a lanostane-type triterpenoid from P. cocos, is a promising bioactive molecule that has demonstrated anti-carcinogenic effects in some tumor models [10, 20–22]. However, the role of PA in lung cancer remains unclear. We therefore undertook a comprehensive analysis of the effects of PA on lung cancer cells using both in vitro and in vivo models. In this study, we demonstrated that PA was effective against lung cancer cells. Our data also showed that PA induced G2/M phase arrest in NCI-H23 and NCI-H460 lung cancer cells. PA-induced G2/M cell cycle arrest provides an opportunity for lung cancer cells to undergo apoptotic progression. Moreover, annexin V/PI double-staining and the DNA ladder formation assays showed that treatment with PA caused concentration-dependent cell apoptosis. These data are consistent with previous report that PA induces apoptosis in prostate cancer [10].

A clearer understanding of how PA regulates the cellular death pathway would provide important insights into the underlying mechanisms. Therefore, we tried to investigate the anti-cancer mechanisms of PA. Mitogen-activated protein kinases (MAPKs) are serine-threonine protein kinases that play major roles in signal transduction from the cell surface to the nucleus. Studies have demonstrated that reactive oxygen species (ROS) are also involved in the regulation of different signal transduction pathways, including MAPKs and transcription factors [23]. In the present study, we found that treating NCI-H23 and NCI-H460 lung cancer cells with PA increased the levels of all three pathways in MAPKs. Surprisingly, only JNK activation is involved in PA-mediated cell death. In addition, considerable evidences suggested that JNK is primarily activated by various environmental factors including oxidative stress [24]. Our data also showed that ROS blockage inhibited PA-induced JNK phosphorylation, indicating that PA-induced apoptosis through ROS-dependent JNK apoptotic pathway. Moreover, it has been well known that activation of JNK pathway induces mitochondria-dependent cell apoptosis via activating mitochondrial Bcl-2 family proteins [25]. We found that PA stimulation significantly increased the level of pro-apoptotic Bax and decreased the level of anti-apoptotic Bcl-2 in NCI-H23 and NCI-H460 cells. Furthermore, we showed that PA-induced activation of Bcl-2/Bax pathway was blocked by ROS inhibitor NAC, indicating that ROS acts as upstream signaling molecules involved in PA-induced activation of mitochondrial pathway.

It is well documented that in response to oxidative stress mediated by ROS, accumulation of unfolded or misfolded proteins triggers a cellular adaptive procedure known as ER stress [26]. Cells initially adapt to the accumulation of unfolded proteins by inducing the expression of ER-resident molecular chaperones as well as by inhibiting nascent protein synthesis [27]. However, if this adaptation does not prove sufficient, the apoptotic response is initiated via induction of ER-associated apoptotic molecules. Among the ER-stress related proteins, ATF4, ATF6 and XBP-1 are typical ER stress-regulated proteins involved in ER stress-induced apoptosis [26, 28]. Previous studies have demonstrated that the regulation of ROS on ER stress activation in a variety of cancer cell lines, including lung cancer cell [29–31]. Additionally, it has been reported that growth inhibition and induction of apoptosis by PA in gemcitabine-resistant pancreatic cancer cells were associated with ER stress activation both in vitro and in vivo [32]. In agreement with previous reports, our findings showed that ATF4, ATF6 and XBP-1 were induced in a dose-dependent manner after PA stimulation, suggesting that ER stress is activated after PA treatment. These initiations of ER stress apoptotic pathway have been reported to increase CHOP gene expression, triggering ER stress-specific cascade for implementation of apoptosis [33]. Consequently, the up-regulation of CHOP protein expression was observed in NCI-H23 and NCI-H460 cells after PA treatment. Moreover, we found that PA-induced activation of ER stress was blocked by ROS inhibitor NAC, indicating that ROS production is also the upstream regulator of PA-induced ER stress in lung cancer cells.

Although in vitro cell culture models are a good system for preliminary screening of the effects of chemotherapeutic agents; the observations must be verified in vivo using animal models, prior to consideration of their potential use in humans. We therefore used an in vivo model of xenografts of NCI-H23 tumor cells in nude mice to verify the chemotherapeutic potential of PA against lung cancer cell growth. Our study provided evidence that administration of PA inhibited the growth of NCI-H23 tumor xenografts without any apparent sign of toxicity in the mice. These data are in accordance with the decreased proliferation documented by Ki67 immunostaining. Additionally, PA induced apoptosis as indicated by TUNEL staining in tumor tissues, at least in part, by activating JNK and ER stress apoptotic pathways. These in vivo results are consistent with our in vitro study, which further suggests that PA may be an applicable approach to enhance the anti-tumor activity against lung cancer.

Conclusion

The present study demonstrates that PA activates ROS-dependent JNK and ER stress apoptotic pathways, and invokes strong anti-cancer activity against lung cancer by inducing apoptosis as well as inhibiting cell growth/proliferation. Thus PA appears to be an potentially attractive bioactive phytochemical for lung cancer treatment.

Methods

Antibodies and reagents

Antibodies specific for p-JNK, JNK, Bcl-2, Bax, Ki67, β-actin and the secondary antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Antibodies specific for CHOP, ATF4, ATF6, p-ERK, ERK, p-p38 and XBP-1 were purchased from Cell Signaling Technology (Danvers, MA). Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) apoptosis detection kit was supplied by Biouniquer Tech (Nanjing, China). SP600125, SB203580, PD98059 and N-acetylcysteine (NAC) were purchased from Sigma (St. Louis, MO). Pachymic acid (Fig. 1a) was purchased from Chengdu Biopurify Phytochemicals Ltd. (Chengdu, Sichuan, China). Other chemicals were obtained from Sigma (St Louis, MO).

Cell culture

The human lung cancer cell lines NCI-H23 and NCI-H460 were purchased from American Type Culture Collection (Rockville, MD), and cultured as recommended as monolayers in DMEM medium (GibcoBRL Life Technologies, Grand Island, NY) supplemented with 10% fetal bovine serum (FBS; GibcoBRL Life Technologies) and 1% penicillin–streptomycin-neomycin (GibcoBRL Life Technologies), in a humidified incubator at 37°C in a 5% CO2 atmosphere.

MTT assay

Cell viability was analyzed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cells were seeded in 96-well plates at a density of 1,000–1,500 cells/well and were treated with PA at the indicated dose for 24, 48 or 72 h. Approximately 20  μl of MTT (5 mg/ml; Sigma, St Louis, MO, USA) was added to each well and incubated for 4 h. At the end of incubation, supernatants were removed, and 150 μl of dimethylsulfoxide (Sigma) was added to each well. The absorbance value (optical density) of each well was measured at 490 nm. All experiments were performed thrice.

Colony formation assay

Cells were plated in 6-well culture plates at 600 cells/well. Each cell group had two wells. Cells were treated with PA at the indicated dose for 24 h. After incubation for another 10 days at 37°C, cells were washed twice with phosphate buffered saline and stained with hematoxylin solution. The number of colonies containing >50 cells was counted under a microscope. The colony formation efficiency was calculated as (number of colonies/number of cells inoculated) × 100%. All assays were independently performed in triplicate.

EdU proliferation assay

The proliferation of NCI-H23 and NCI-H460 cells were examined using the Cell-Light EdU Apollo488 In vitro Imaging Kit (RiboBio) according to the manufacturer’s protocol. Briefly, cells were incubated with 10 μM EdU for 2 h before fixation with 4% paraformaldehyde, permeabilization by 0.3% Triton X-100 and EdU staining. Cell nuclei were stained with 5 μg/ml DAPI (4′,6-diamidino-2-phenylindole) for 10 min. The number of EdU-positive cells was counted under a microscope in five random fields (×100). All assays were independently performed in triplicate.

Cell cycle and apoptosis assay

Flow cytometric analysis was performed to determine the presence of cell cycle arrest and apoptotic cells. After treatment with PA for 24 h, the cells were collected by trypsinisation and washed twice with PBS, fixed in ice-cold 80% ethanol, and stored overnight at 4°C. For analysis, the cells were washed twice with PBS and 10 mg/ml RNase A was added. Propidium iodide was added to the tubes at a final concentration of 0.05 mg/ml and incubated at 4°C for 20 min in the dark. Cell cycle analysis was performed with FACSCalibur flow cytometer (BD Biosciences, CA). FITC-labeled Annexin V/PI staining was performed according to the manufacturer’s instructions (Keygen, Nanjing, China). Briefly, 1 × 106 cells/well were suspended in buffer containing FITC-conjugated Annexin V/PI at appropriate concentrations. The samples were analyzed by flow cytometry and 20,000 events from each sample were obtained to ensure adequate data.

DNA fragment analysis

The cells (1.5 × 106) were treated with PA (20, 40 or 80 µM) for 24 h, then collected and washed twice with PBS. DNA was extracted using the Genomic DNA Mini Preparation kit with Spin Column (Beyotime Institute of Biotechnology, Haimen, China). DNA (~15 μg) was loaded onto a 1.5% agarose gel. Subsequent to electrophoresis, the gel was visualized using ethidium bromide staining under ultraviolet light.

Western blotting

Cells were seeded in 6-well plates at a density of 1 × 106 cells/well with 2 ml completed DMEM medium. Following PA treatment for the indicated times, total protein was extracted. The protein concentration was determined using the BCA method. Equal quantities of proteins were separated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and transferred by electroblotting onto a nitrocellulose membrane. Membranes were incubated with the primary antibody at 4°C overnight. After additional TBST washes, membranes were incubated with corresponding horseradish peroxidase-conjugated secondary antibodies (Bio-Rad) for 1 h at room temperature and detected by the enhanced chemiluminescence method (SuperSignal West Pico substrate; Pierce; Rockford, IL).

Tumor xenograft model

All procedures and experiments involving animals in this study were performed in accordance with the National Institutes of Health Guide for Care and Use of Laboratory Animals. The study protocol was approved by the Animal Ethics Committee at Fudan University, China. Female athymic nude mice of 4–5 weeks of age were purchased from Shanghai SLAC Laboratory Animal Co., Ltd. (Shanghai, China) and were housed in the Animal Resource Facility. Exponentially growing NCI-H23 cells (5 × 106 in 100 µl PBS) were injected subcutaneously in the right flank of each mouse. Tumor xenografts were allowed to grow to an average size of 100–200 mm3 and were randomly assigned to four different treatment groups (six mice per group): (a) vehicle control (0.1% DMSO in physiological saline); (b) PA 10 mg/kg; (c) PA 30 mg/kg; (Dd) PA 60 mg/kg. The mice were administered PA via intraperitoneal (ip) injections for 3 weeks (5 days/week). Tumor size was measured on two axes with the aid of Vernier calipers and tumor volume (mm3) was calculated using the formula: 1/2(L × W2) where L is the longest and W is the shortest axis. Mice were euthanized at the end of the study and/or when tumor size exceeded 2,000 mm3. At the end of the experiments, the xenograft tumors, lungs, livers, kidneys and spleens were removed from the mice and snap-frozen for immunohistochemistry and hematoxylin and eosin staining.

TUNEL assay for apoptotic cells

The type of cell death (necrosis/apoptosis) was evaluated by the terminal deoxynucleotidyl transferase-mediated deoxyuridine biotin nick-end labeling (TUNEL) assay with an Apo-Direct kit (Pharmingen, San Diego, CA). Briefly, after antigen retrieval, the tumor sections (4 µm-thick) were fixed by incubation with 4% paraformaldehyde at 4°C. The permeabilized sections were incubated with terminal deoxynucleotidyl transferase recombinant (rTdT) enzyme-catalysed reaction and nucleotide mixture for 60 min at 37°C in the dark. After immersion in stop/wash buffer for 15 min at room temperature, the sections were washed with PBS to remove unincorporated fluorescein-12-dUTP and the nuclei counterstained with hematoxylin.

Hematoxylin and eosin (H&E) staining and immunohistochemistry

The xenograft tumors, lungs, livers, kidneys and spleens were performed hematoxylin and eosin staining. Briefly, freshly dissected tissues were fixed and embedded in paraffin. After being cut into 4-μm slices, the sections were deparaffinized and stained in Mayer’s Hematoxylin and Eosin solution. Finally, the sections were dehydrated and mounted with Permount in a fume hood. The results were analyzed under a phase-contrast Olympus microscope (Olympus America Inc). For the immunohistochemical studies, the freshly dissected tumor tissue was fixed in 10% neutral buffered formalin for 24–48 h. The tissue block was embedded in paraffin and cut to the desired thickness using a microtome, and was affixed onto a slide. After several wash cycles, the tumor sections were blocked and incubated with antibodies against Ki67, p-JNK and CHOP (diluted 1:200 in 5% horse serum in PBS) overnight at 4°C. Subsequently, sections were incubated with pre-diluted streptavidin-peroxidase HRP conjugates in a humidified chamber at room temperature using a staining kit, according to the manufacturer’s instructions (Dako North America, Inc., CA). The sections were counterstained with hematoxylin for 2–3 min and mounted and analyzed under a phase-contrast Olympus microscope (Olympus America Inc).

Statistical analysis

Data are presented as mean ± SD unless otherwise indicated. The statistical significance of the difference between the values of control and treatment groups was determined by either Student t test or simple one-way ANOVA followed by Tukey’s post hoc test for multiple comparisons using Prism version 5 (GraphPad Software, Inc.). Values of p < 0.05 were considered statistically significant.

Abbreviations

PA:

pachymic acid

ROS:

reactive oxygen species

JNK:

c-Jun N-terminal kinase

ER:

endoplasmic reticulum

References

  1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D (2011) Global cancer statistics. CA Cancer J Clin 61(2):69–90

    Article  PubMed  Google Scholar 

  2. Wang L, Xiong Y, Sun Y, Fang Z, Li L, Ji H et al (2010) Lung DB: an integrated database of human lung cancer research. Nucleic Acids Res 38(Database issue):D665–D669

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Fruh M (2011) The search for improved systemic therapy of non-small cell lung cancer—what are today’s options? Lung Cancer 72(3):265–270

    Article  CAS  PubMed  Google Scholar 

  4. Feng Y, Wang N, Zhu M, Feng Y, Li H, Tsao S (2011) Recent progress on anticancer candidates in patents of herbal medicinal products. Recent Pat Food Nutr Agric 3(1):30–48

    Article  PubMed  Google Scholar 

  5. Elmore S (2007) Apoptosis: a review of programmed cell death. Toxicol Pathol 35(4):495–516

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Fesik SW (2005) Promoting apoptosis as a strategy for cancer drug discovery. Nat Rev Cancer 5(11):876–885

    Article  CAS  PubMed  Google Scholar 

  7. Ocker M, Hopfner M (2012) Apoptosis-modulating drugs for improved cancer therapy. Eur Surg Res 48(3):111–120

    Article  CAS  PubMed  Google Scholar 

  8. Shrotriya S, Deep G, Gu M, Kaur M, Jain AK, Inturi S et al (2012) Generation of reactive oxygen species by grape seed extract causes irreparable DNA damage leading to G2/M arrest and apoptosis selectively in head and neck squamous cell carcinoma cells. Carcinogenesis 33(4):848–858

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Zhou Y, Zhao W, Xie G, Huang M, Hu M, Jiang X et al (2014) Induction of Nur77-dependent apoptotic pathway by a coumarin derivative through activation of JNK and p38 MAPK. Carcinogenesis 35(12):2660–2669

    Article  PubMed  Google Scholar 

  10. Gapter L, Wang Z, Glinski J, Ng KY (2005) Induction of apoptosis in prostate cancer cells by pachymic acid from Poria cocos. Biochem Biophys Res Commun 332(4):1153–1161

    Article  CAS  PubMed  Google Scholar 

  11. Kaminaga T, Yasukawa K, Kanno H, Tai T, Nunoura Y, Takido M (1996) Inhibitory effects of lanostane-type triterpene acids, the components of Poria cocos, on tumor promotion by 12-O-tetradecanoylphorbol-13-acetate in two-stage carcinogenesis in mouse skin. Oncology 53(5):382–385

    Article  CAS  PubMed  Google Scholar 

  12. Ukiya M, Akihisa T, Tokuda H, Hirano M, Oshikubo M, Nobukuni Y et al (2002) Inhibition of tumor-promoting effects by poricoic acids G and H and other lanostane-type triterpenes and cytotoxic activity of poricoic acids A and G from Poria cocos. J Nat Prod 65(4):462–465

    Article  CAS  PubMed  Google Scholar 

  13. Huang YC, Chang WL, Huang SF, Lin CY, Lin HC, Chang TC (2010) Pachymic acid stimulates glucose uptake through enhanced GLUT4 expression and translocation. Eur J Pharmacol 648(1–3):39–49

    Article  CAS  PubMed  Google Scholar 

  14. Kitano H, Chung JY, Ylaya K, Conway C, Takikita M, Fukuoka J et al (2014) Profiling of phospho-AKT, phospho-mTOR, phospho-MAPK and EGFR in non-small cell lung cancer. J Histochem Cytochem 62(5):335–346

    Article  PubMed Central  PubMed  Google Scholar 

  15. Li Y, Li T, Miao C, Li J, Xiao W, Ma E (2013) Beta-Eudesmol induces JNK-dependent apoptosis through the mitochondrial pathway in HL60 cells. Phytother Res 27(3):338–343

    Article  CAS  PubMed  Google Scholar 

  16. Liu Z, Sun Y, Ren L, Huang Y, Cai Y, Weng Q et al (2013) Evaluation of a curcumin analog as an anti-cancer agent inducing ER stress-mediated apoptosis in non-small cell lung cancer cells. BMC Cancer 13:494

    Article  PubMed Central  PubMed  Google Scholar 

  17. Qu K, Shen NY, Xu XS, Su HB, Wei JC, Tai MH et al (2013) Emodin induces human T cell apoptosis in vitro by ROS-mediated endoplasmic reticulum stress and mitochondrial dysfunction. Acta Pharmacol Sin 34(9):1217–1228

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Zou P, Zhang J, Xia Y, Kanchana K, Guo G, Chen W et al (2015) ROS generation mediates the anti-cancer effects of WZ35 via activating JNK and ER stress apoptotic pathways in gastric cancer. Oncotarget 6:5860–5876

    PubMed Central  PubMed  Google Scholar 

  19. Newman DJ, Cragg GM, Snader KM (2003) Natural products as sources of new drugs over the period 1981–2002. J Nat Prod 66(7):1022–1037

    Article  CAS  PubMed  Google Scholar 

  20. Hong R, Shen MH, Xie XH, Ruan SM (2012) Inhibition of breast cancer metastasis via PITPNM3 by pachymic acid. Asian Pac J Cancer Prev 13(5):1877–1880

    Article  PubMed  Google Scholar 

  21. Ling H, Jia X, Zhang Y, Gapter LA, Lim YS, Agarwal R et al (2010) Pachymic acid inhibits cell growth and modulates arachidonic acid metabolism in nonsmall cell lung cancer A549 cells. Mol Carcinog 49(3):271–282

    CAS  PubMed  Google Scholar 

  22. Ling H, Zhang Y, Ng KY, Chew EH (2011) Pachymic acid impairs breast cancer cell invasion by suppressing nuclear factor-kappaB-dependent matrix metalloproteinase-9 expression. Breast Cancer Res Treat 126(3):609–620

    Article  CAS  PubMed  Google Scholar 

  23. Ray PD, Huang BW, Tsuji Y (2012) Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal 24(5):981–990

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Zhou H, Shen T, Shang C, Luo Y, Liu L, Yan J et al (2014) Ciclopirox induces autophagy through reactive oxygen species-mediated activation of JNK signaling pathway. Oncotarget 5(20):10140–10150

    PubMed Central  PubMed  Google Scholar 

  25. Lei K, Davis RJ (2003) JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis. Proc Natl Acad Sci USA 100(5):2432–2437

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Boyce M, Yuan J (2006) Cellular response to endoplasmic reticulum stress: a matter of life or death. Cell Death Differ 13(3):363–373

    Article  CAS  PubMed  Google Scholar 

  27. Schroder M, Kaufman RJ (2005) ER stress and the unfolded protein response. Mutat Res 569(1–2):29–63

    Article  PubMed  Google Scholar 

  28. Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K (2001) XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 107(7):881–891

    Article  CAS  PubMed  Google Scholar 

  29. Moon DO, Park SY, Choi YH, Ahn JS, Kim GY (2011) Guggulsterone sensitizes hepatoma cells to TRAIL-induced apoptosis through the induction of CHOP-dependent DR5: involvement of ROS-dependent ER-stress. Biochem Pharmacol 82(11):1641–1650

    Article  CAS  PubMed  Google Scholar 

  30. Shi JM, Bai LL, Zhang DM, Yiu A, Yin ZQ, Han WL et al (2013) Saxifragifolin D induces the interplay between apoptosis and autophagy in breast cancer cells through ROS-dependent endoplasmic reticulum stress. Biochem Pharmacol 85(7):913–926

    Article  CAS  PubMed  Google Scholar 

  31. Yang KM, Kim BM, Park JB (2014) omega-Hydroxyundec-9-enoic acid induces apoptosis through ROS-mediated endoplasmic reticulum stress in non-small cell lung cancer cells. Biochem Biophys Res Commun 448(3):267–273

    Article  CAS  PubMed  Google Scholar 

  32. Cheng S, Swanson K, Eliaz I, McClintick JN, Sandusky GE, Sliva D (2015) Pachymic acid inhibits growth and induces apoptosis of pancreatic cancer in vitro and in vivo by targeting ER stress. PLoS One 10(4):e0122270

    Article  PubMed Central  PubMed  Google Scholar 

  33. Marciniak SJ, Yun CY, Oyadomari S, Novoa I, Zhang Y, Jungreis R et al (2004) CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev 18(24):3066–3077

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Authors’ contributions

JM and DFC participation in study designed; JM, JL, and CWL conducting the study in the labs; JM and DFC Data analyzing and manuscript preparation. All authors read and approved the final manuscript.

Acknowledgements

This study was supported by The National Natural Science Foundation of China (Grant no. 81201897; 81201912), The Program of Fudan University improving young teacher’s research ability (Grant no. 20520133503).

Compliance with ethical guidelines

Competing interests The authors declare that they have no competing interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dingfang Cai.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, J., Liu, J., Lu, C. et al. Pachymic acid induces apoptosis via activating ROS-dependent JNK and ER stress pathways in lung cancer cells. Cancer Cell Int 15, 78 (2015). https://doi.org/10.1186/s12935-015-0230-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-015-0230-0

Keywords