Oridonin inhibits growth and induces apoptosis of human neurocytoma cells via the Wnt/β‑catenin pathway

  • Authors:
    • Jingyan Liang
    • Weiguang Wang
    • Lifu Wei
    • Shan Gao
    • Yingge Wang
  • View Affiliations

  • Published online on: June 18, 2018     https://doi.org/10.3892/ol.2018.8977
  • Pages: 3333-3340
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Central neurocytoma (CN) is a rare periventricular tumor of the central nervous system in young adults. Typically, patients with CN exhibit a favorable prognosis, but in certain cases the clinical course is more aggressive. Therefore, investigating effective therapeutic approaches is important. Oridonin has attracted attention due to its antitumor activities. However, the role of oridonin in tumorigenesis and progression remains unknown. The present study examined the antitumor function of oridonin in CN cells, and investigated the underlying molecular mechanism. An MTT assay suggested that treatment with oridonin was able to significantly inhibit the proliferation of CN cells. The annexin V‑fluorescein isothiocyanate/propidium iodide assay and western blot analysis demonstrated that oridonin was able to induce apoptosis and alter the expression of apoptosis‑associated proteins by downregulating anti‑apoptotic protein, B‑cell lymphoma‑2 (Bcl‑2), and upregulating pro‑apoptosis proteins, Bcl‑2‑like protein 4, cleaved caspase‑3 and cleaved poly(ADP‑ribose) polymerase 1. Subsequently, the Wnt/β‑catenin signaling pathway was examined. Western blot analysis indicated that oridonin markedly decreased the expression of β‑catenin, cyclin D1 and v‑myc avian myelocytomatosis viral oncogene homolog. Furthermore, β‑catenin was silenced by small interference RNA or overexpressed in CN cells, and the effect on cell proliferation was examined. The results indicated that silencing of β‑catenin enhanced the inhibitory effect of oridonin on cell growth, whereas the overexpression of β‑catenin attenuated this effect. These data indicated that oridonin inhibited proliferation and induced apoptosis to exert its antitumor activity in CN cells by repressing Wnt/β‑catenin signaling. Therefore, the present study suggested that oridonin might be an effective adjuvant agent, and that the Wnt/β‑catenin signaling pathway may be a potent target for the therapy in CN.

Introduction

Central neurocytoma (CN) is a rare but usually benign neuronal tumor composed of uniform round cells with neuronal differentiation and typically located in the supratentorial ventricles in young adults (1,2). Surgery and radiotherapy are widely considered as the most important therapeutic approaches for CN. Patients with CN commonly exhibit a good prognosis, but in certain cases the clinical course is more aggressive or is followed by recurrence (3). Chemotherapy may be useful for recurrent CN that cannot be resected and has been radiated. Therefore, it is necessary to develop effective antitumor drugs for adjuvant therapy of CN. In particular, the combination of antitumor drugs with molecular target therapy may greatly improve the prognosis of CN.

Oridonin, a diterpenoid isolated from the Chinese medicinal herb Rabdosia rubescens, exhibits potential anti-inflammatory, anti-tumor, pro-apoptotic and neurological effects (4,5). Oridonin has been demonstrated to inhibit proliferation and induce apoptosis in a variety of human cancer cells, including colon, pancreatic, breast, lung and liver cancer (610). However, the underlying mechanisms remain poorly understood. Previously, oridonin has been demonstrated to exert antitumor activities through several signaling pathways that are associated with cell proliferation and apoptosis, including c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase (ERK) and protein kinase B (Akt) signaling pathways (11,12). Although oridonin exhibits a significant antitumor function in multiple types of cancer, its exact effect on CN and the underlying mechanism remain unclear.

The Wnt/β-catenin signaling pathway is a canonical Wnt pathway, which serves a critical role in regulating cell growth, apoptosis, motility, polarity and differentiation (13). Dysregulation of the pathway is identified in several types of cancer, including liver, colon and several types of brain tumors (14). The underlying molecular mechanisms of CN have been largely examined, but it has been suggested that the receptors and effectors of the Wnt pathway are differentially overexpressed in CN cells (15). Additionally, it has also been demonstrated that oridonin inhibits the proliferation of human osteosarcoma cells by suppressing Wnt signaling (16). Therefore, the present study hypothesized whether oridonin may inhibit the growth of CN cells by affecting Wnt/β-catenin signaling transduction.

In the present study, the roles of oridonin in the proliferation and apoptosis of CN cells as well as the potential molecular mechanisms were investigated. It was indicated that oridonin was able to inhibit proliferation and induce apoptosis, which might be mediated by repressing the Wnt/β-catenin signaling pathway in CN cells.

Materials and methods

Cell culture and treatment

Central neurocytoma tissue was obtained from 1 patient diagnosed with CN for cell culture following resection at the First Affiliated Hospital of Jiamusi University (Jiamusi, China). Resected tissues were stored in Mg2+/Ca2+-free Hank's Balanced Salt Solution (HBSS), and then cut into small pieces in piperazine-N,N′-bis(2-ethanesulfonic acid) (PIPES) solution (20 mM PIPES, 25 mM glucose, 5 mM KCl, 120 mM NaCl) and treated as previously described (1719). The tissue samples were resuspended in Dulbecco's modified Eagle's medium/F-12/N2 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 5% fetal bovine serum (FBS; Lonza Group, Ltd., Basel, Switzerland), 100 U/ml penicillin, 100 µg/ml streptomycin, 10 ng/ml epidermal growth factor (EGF) and 20 ng/ml basic fibroblast growth factor (bFGF). A total of 1×106 dissociated cells/well were plated into collagen IV (BD Biosciences, Franklin Lakes, NJ, USA) or fibronectin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) precoated 6-well culture plate. The cells were cultured in a 37°C incubator with 5% CO2. The present study design was approved by the Ethics Committee of First Affiliated Hospital of Jiamusi University, and all patients provided written informed consent.

RNA interference and overexpression of β-catenin

Small interfering RNA (siRNA) targeting β-catenin (siβ-catenin) and recombinant adenoviruses expressing β-catenin (Ad-β-ctn) were obtained as previously described using the AdEasy technology (2022). The sequence of β-catenin siRNA was 5′-CAGGGGGUUGUGGUUAAGCUCUU-3′. A scramble siRNA sequence (5′-TTCTCCGAACGTGTCACGT-3′) was used as a control (Gima Biol Engineering Inc., Shanghai, China). A total of 2×104 cultured central neurocytoma cells were seeded into each well of a 12-well plate and were cultured to 80% confluence. Cell transfections were performed using 100 nmol siRNA and 5 µl Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Cells were further cultured for 48 h following transfection, and cells were subsequently lysed and analyzed for the protein expression of β-catenin by western blotting. In addition, cells were treated with oridonin, and then cell proliferation was detected.

Proliferation assay

Cell proliferation was assessed by MTT assay. Briefly, following transfection, 104 cells were seeded in a 96-well flat bottom plate. The cells were cultured in a 5% CO2 incubator at 37°C. Once cell confluence reached 80%, the supernatant was replaced with fresh medium and the cells were treated with 0, 5, 10, 15, 20 or 25 µM oridonin (Xi'an Hao-Xuan Bio-tech Co., Ltd., Xi'an, China) dissolved in dimethyl sulfoxide (DMSO) for 24, 48 or 72 h followed by an additional 4 h subsequent to the addition of 20 µl MTT (5 mg/ml) into each well. A total of 200 µl DMSO was added to the wells for cell lysis. Absorbance was detected using an ELISA spectrophotometer at 490 nm.

Apoptosis assay

A total of 1×106 central neurocytoma cells were seeded on 60 mm dishes and cultured in Dulbecco's modified Eagle's/Nutrient Mixture F12 (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 5% fetal bovine serum (Lonza Group, Ltd., Basel, Switzerland), 100 U/ml penicillin and 100 µg/ml streptomycin. When cells reached 80% confluence, they were treated with 0, 10, 15 or 20 µM oridonin for the indicated time. Apoptosis was quantified by Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) assay (BD Biosciences, Franklin Lakes, NJ, USA) following the manufacturer's protocol. Briefly, cells were collected by trypsinization and pooled with cell supernatants from corresponding culture dishes; 500 µl of this cell suspension was incubated with 2 µl of Annexin-V-FITC stock solution for 15 min in the dark. After a short centrifugation (300 × g, 5 min), cells were washed in PBS before being resuspended in 0.5 ml of 1× binding buffer supplemented with 1 µl PI (1 µg/ml final concentration). The Annexin V-FITC/PI assay detects the amount of phosphatidylserine on the outer surface of the plasma membrane (a biochemical alteration unique to membranes of apoptotic cells) and the amount of PI, a dye that easily enters dead cells or cells in the late stages of apoptosis and binds DNA but does not bind with the plasma membrane of viable cells. Fluorescence was detected using a FACSCalibur flow cytometer by fluorescence activated cell sorter (FACS) analysis, and data were analyzed using CellQuestPro version 5.2 software (BD Biosciences, San Jose, CA, USA). The cells with phosphatidylserine on their surface were considered to be apoptotic.

Western blot analysis

The cells were washed twice with PBS and were lysed with lysis buffer (50 mM Tris-HCl (pH 7.4), 1 mM EDTA, 1% NP40, 150 mM NaCl, 10 mM NaF and 1 mM Na3VO4) containing a protease inhibitor cocktail (Roche Molecular Diagnostics, Branchburg, NJ, USA). Following centrifugation at 12,000 × g for 10 min at 4°C, the supernatant was collected and quantified using a bicinchoninic acid quantification kit (Beyotime Institute of Biotechnology, Haimen, China). 50 µg proteins were loaded per lane onto 10% SDS-PAGE gels (Bio-Rad Laboratories, Inc., Hercules, CA, USA) and transferred to Immobilon-P membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked with 5% skimmed milk in Tris-buffered saline containing 0.05% Tween 20 for 1 h at room temperature, and incubated with the following specific primary antibodies overnight at 4°C. The antibodies against B-cell lymphoma-2 (Bcl-2; 1:500; catalogue no. sc7382; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), Bcl-2-like protein 4 (Bax; 1:500; cat no. sc493; Santa Cruz Biotechnology, Inc.), cleaved caspase-3 (1:1,000; cat no. 9661S; Cell Signaling Technology, Inc., Danvers, MA, USA) and cleaved poly (ADP-ribose) polymerase 1 (cleaved PARP; 1:1,000; catalogue no. 9541; Cell Signaling Technology, Inc.), β-catenin (1:500; catalogue no. sc1496; Santa Cruz Biotechnology, Inc.), cyclin D1 (1:1,000; catalogue no. sc20044; Santa Cruz Biotechnology, Inc.), v-myc avian myelocytomatosis viral oncogene homolog (c-Myc; 1:1,000; catalogue no. sc788; Santa Cruz Biotechnology, Inc.) and β-actin (1:1,000; catalogue no. sc47778; Santa Cruz Biotechnology, Inc.) were used for detection. This was followed by incubation with horseradish peroxidase-conjugated goat anti-mouse secondary antibody (1:2,000; catalogue no. sc-2005; Santa Cruz Biotechnology, Inc.) and anti-rabbit immunoglobulin G antibody (1:2,000; catalogue no. sc-2004; Santa Cruz Biotechnology, Inc.) for 2 h at room temperature. The blots were visualized using enhanced chemiluminescence detection reagent (GE Healthcare Life Sciences, Little Chalfont, UK). The gray value of the targeted bands was quantified with QuantityOne software version 4.6.2 (Bio-Rad Laboratories, Inc., Hercules, CA, USA) following incubation, with β-actin used as the internal reference.

Quantitative reverse-transcriptase polymerase chain reaction (RT-qPCR)

Total RNA was extracted from cells using TRIzol reagent according to the manufacturer's protocol (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). cDNA was generated with reverse transcription using the RevertAid First Strand cDNA synthesis kit (Fermentas, Thermo Fisher Scientific, Inc., Pittsburgh, PA, USA) and was amplified using a TaqMan® Gene Expression Assay (Applied biosystems; Thermo Fisher Scientific, Inc.) with fluorogenic carboxyfluorescein-labeled probes using specific primers for target proteins. The specific primers for PCR were forward, 5′-ACCAGTGGATTCTGTGTTGTT-3′ and reverse, 5′-ATTTGAAGGCAGTCTGTCGTA-3′ for β-catenin and forward, 5′-GATCCCTCCAAAATCAAGTG-3′ and reverse, 5′-GAGTCCTTCCACGATACCAA-3′ for GAPDH. Real-time fluorescence detection was performed with the ABI PRISM 7700 Sequence Detector (Applied Biosystems; Thermo Fisher Scientific, Inc.). PCR involved 40 amplification cycles of 94°C for 10 sec, 53°C for 30 sec and 72°C for 40 sec, followed by final extension at 72°C for 10 min. β-catenin expression was normalized to GAPDH expression and calculated using the 2−ΔΔCt formula (23). The relative level of β-catenin mRNA was presented as a percentage of the control.

Statistical analysis

All of the experiments were repeated at least 3 times. SPSS (version 16.0; SPSS, Inc., Chicago, IL, USA) was used to analyze the experimental data. One-way analysis of variance was used to assess the differences between the groups. Duncan's multiple range test was employed for pairwise comparison and followed by Bonferroni correction. The data are presented as the mean ± standard error of the mean. P<0.05 (two-tailed) was considered statistically significant difference.

Results

Oridonin suppresses the proliferation of CN cells

To determine the role of oridonin in CN cells, CN tissue was obtained from one patient diagnosed with CN following resection, and a CN cell line was generated by primary culture (1719). The cells were grown in the medium for 24 h, and then treated with 10, 15 and 20 µM oridonin for 48 h or were treated with 5, 10, 15, 20 or 25 µM oridonin for 24, 48 or 72 h. An MTT assay was performed to evaluate cell proliferation. The results demonstrated that oridonin was able to markedly decrease the proliferation of CN cells in a concentration- and time-dependent manner relative to the DMSO-treated control (Fig. 1A and B).

Oridonin induces the apoptosis of CN cells

To confirm whether the inhibition of proliferation that is mediated by oridonin is associated with cell apoptosis, the cells were treated with 10, 15 and 20 µM oridonin for 48 h. Apoptosis was quantified by FACS analyses. The results demonstrated that oridonin treatment was able to significantly promote the apoptosis of CN cells in a concentration-dependent manner (Fig. 2A). Additionally, CN cells were treated with 15 µM oridonin for 24, 48 and 72 h. It was identified that apoptosis was induced by oridonin in a time-dependent manner (Fig. 2B). These results were consistent with the pattern observed with the inhibition of proliferation, suggesting that oridonin acted as a potent apoptotic inducer in CN cells.

Oridonin affects the expression of apoptosis-associated proteins in CN cells

To additionally determine the role of oridonin in the apoptosis of CN cells, western blotting was performed to assess the expression levels of apoptosis-associated proteins: Bcl-2, Bax, cleaved caspase-3 and cleaved PARP. The results demonstrated that the expression of Bcl-2, an anti-apoptotic protein, was significantly reduced in oridonin-treated CN cells compared with control cells. By contrast, the expression levels of pro-apoptosis protein Bax, cleaved caspase-3 and cleaved PARP were increased (Fig. 3A and B), and the Bax/Bcl-2 ratio was increased (Fig. 3C). The Bax/Bcl-2 ratio is a key factor in determining the occurrence and level of apoptosis (24). These results indicated that treatment with oridonin was able to alter cell apoptosis, potentially by modulating Bcl-2, Bax, and cleaved caspase-3 and cleaved PARP.

Oridonin downregulates the Wnt/β-catenin signaling pathway

To investigate the molecular mechanism underlying the effect of oridonin on the growth and apoptosis of CN cells, the Wnt/β-catenin signaling pathway was evaluated by detecting the expression of key proteins of this pathway. CN cells were treated with 15 µM oridonin for 48 h. Western blot analysis was performed, and the results indicated that treatment with oridonin was able to downregulate the accumulation of β-catenin, cyclin D1 and c-Myc in CN cells. Densitometric analysis of the western blot bands confirmed these results (Fig. 4A and B), suggesting that oridonin regulated the growth and apoptosis of CN cells, which is likely to be mediated by Wnt signaling.

Silencing and overexpressing β-catenin in CN cells

To verify the involvement of Wnt/β-catenin signaling pathway in oridonin-mediated inhibition on proliferation of CN cells, β-catenin was knocked down using siRNA or was overexpressed by infecting cells with recombinant adenoviruses that expressed β-catenin in CN cells. The RNA expression level of β-catenin was examined by RT-qPCR, and protein expression was detected by western blot analysis. The results indicated that the relative β-catenin mRNA expression was markedly decreased in cells that were treated with β-catenin siRNA and the residual protein expression of β-catenin in the cells was markedly reduced compared with the control siRNA-treated cells (Fig. 5A and B). Additionally, western blot analysis indicated that β-catenin was significantly elevated in cells that overexpressed β-catenin (Fig. 5C).

Role of β-catenin in oridonin-mediated inhibition of cell proliferation

The effect of β-catenin, a key member of the Wnt signaling pathway, on oridonin-mediated inhibition of cell proliferation in CN cells was examined in the aforementioned cell lines. β-catenin-silenced and non-silenced CN cells were treated with or without 15 µM oridonin for 48 h. Detection of cell proliferation indicated that treatment with oridonin and silencing of β-catenin inhibited cell proliferation and induced apoptosis (Fig. 6A). The combination of oridonin treatment and silencing of β-catenin markedly augmented the effects on cell proliferation and apoptosis compared with oridonin single treatment (Fig. 6A). Additionally, the overexpression of β-catenin attenuated the effects of oridonin on proliferation and apoptosis (Fig. 6B). These data suggested that the Wnt/β-catenin signaling pathway served an important role in the antitumor activities of oridonin in CN cells, and that oridonin regulated the growth of CN cells, which might be via the Wnt/β-catenin signaling pathway.

Discussion

Oridonin is a diterpenoid compound isolated from the Chinese traditional medicine herb Rabdosia rubescens (25), which has attracted attraction due to its antitumor activities (26). Oridonin has been demonstrated to exert antitumor effects by inhibiting cell growth, proliferation and inducing apoptosis in multiple types of human cancer (27). However, the role of oridonin in the regulation of biological function of CN cells and the underlying molecular mechanisms remain unclear. The molecular regulatory mechanisms in CN cells are predominantly unexplored. In the present study, the data suggested that oridonin may suppress cell proliferation and induce apoptosis in CN cells, and that the function may be mediated by altering the Wnt/β-catenin signaling pathway. To investigate the molecular mechanisms underlying the inhibition of cell proliferation and induction of apoptosis in CN cells, the expression levels of apoptosis-associated proteins, Bcl-2, Bax, cleaved caspase-3 and cleaved PARP, were detected by western blotting in the present study.

Anti-apoptotic Bcl-2 and pro-apoptotic Bax as well as Bcl-2 family proteins regulate mitochondrial permeability to alter apoptosis via an intrinsic pathway (28). In the present study, treatment with oridonin was able to decrease Bcl-2 expression and increase Bax expression in CN cells. Furthermore, the Bax/Bcl-2 ratio (an important parameter to measure the occurrence and levels of apoptosis) was significantly elevated.

Cleaved caspase-3, also known as mature or activated caspase-3, is a critical mediator of cell apoptosis (29). Pro-caspases require cleavage after aspartic acid residues, which result in one large and one small subunit. These subunits associate into an a2b2 tetramer to form the active enzyme (30,31). Caspase-3 is able to cleave PARP to a specific 85-kDa form, which is observed during apoptosis (30,31). In the present study, treatment with oridonin was able to increase the levels of cleaved caspase-3 and cleaved PARP, which was consistent with the promotion of apoptosis. These data indicated that oridonin-induced apoptosis in CN cells is associated with a decrease in Bcl-2 expression and an increase in Bax expression and activation of caspase-3.

The canonical Wnt signaling pathway serves an important role in the regulation of cell proliferation and apoptosis. It has been indicated that downstream target genes of the Wnt/β-catenin pathway, including c-Myc and cyclin D1, are associated with apoptosis (32,33). The Wnt signaling pathway is activated by interactions between the Wnt ligand and the Frizzled family receptor and the low-density lipoprotein receptor-related protein 5/6. The interaction leads to an accumulation of β-catenin in the cytoplasm and translocation into the nucleus due to the inactivation of the β-catenin destruction complex, Axin/adenomatous polyposis coli/glycogen synthase kinase-3β (3437). Subsequently, β-catenin interacts with TCF4/LEF to activate the transcription of the target genes Bcl-2, cyclin D1 and c-Myc, which control the transition from G1 to S, resulting in abnormal cellular proliferation and apoptosis (3840). The deregulation of Wnt signaling has been identified to be involved in tumorigenesis and the development of various types of cancer (41). It has been demonstrated that the Wnt pathway receptor, Frizzled-1, and the effector, T cell transcription factor 4 (TCF4), are highly expressed in CN cells and involved in the origin and expansion of neurocytoma from native subependymal progenitor cells (15), suggesting that Wnt/β-catenin signaling is activated in CN cells. Combined with the reported finding that oridonin inhibits Wnt signaling in osteosarcoma cells (16), it is possible that oridonin may affect Wnt signaling in CN cells.

The stability of β-catenin is commonly used to evaluate the activity of the Wnt/β-catenin signaling pathway. In the present study, it was revealed that oridonin was able to downregulate the level of β-catenin protein in CN cells in a concentration- and time-dependent manner. It has been previously reported that β-catenin is able to bind TCFs to stimulate cellular growth and proliferation in tumorigenesis by triggering the cell cycle regulator cyclin D1 (42). c-Myc, as the target of β-catenin protein, also serves a critical role in tumor prognosis (43). In the present study, it was demonstrated that cyclin D1 and c-Myc, the downstream targets of β-catenin, were reduced in CN cells, indicating that the Wnt/β-catenin pathway was inhibited. Additionally, silencing β-catenin was able to augment oridonin-mediated inhibition of proliferation, whereas the overexpression of β-catenin was able to attenuate these effects in CN cells. These findings indicated that the antitumor activity of oridonin is mediated via the Wnt/β-catenin signaling pathway in CN cells.

The potential molecular mechanism underlying the antitumor activities of oridonin has been investigated. Several studies indicated that certain signaling pathways and key genes associated with cell apoptosis and cells cycle were regulated by oridonin (44). For instance, oridonin is able to induce autophagy and apoptosis by upregulating p21 in prostate cancer cells (43) or can downregulate the phosphoinositide 3-kinase/Akt signaling pathway to suppress proliferation and induce caspase-dependent apoptosis in cervical cancer HeLa cells (45). In addition, oridonin may downregulate the activities of ERK and Akt and stimulate c-Jun and MAPK pathways to suppress proliferation and induce apoptosis in osteosarcoma cells (46). Oridonin has also been suggested to suppress the protein tyrosine kinase-Ras-Raf-JNK survival pathway and activate the ERK-p53 apoptotic pathway, resulting in cell cycle arrest and apoptosis in murine fibrosarcoma cells (47). Therefore, whether other signaling pathways or proteins take part in the regulation of antitumor activities of oridonin in CN cells remains to be elucidated.

In conclusion, the results of the present study indicated that oridonin exerts its antitumor function in CN cells by downregulating the activity of the Wnt/β-catenin signaling pathway, suggesting that oridonin and other compounds from Chinese herbal medicines targeting the Wnt/β-catenin signaling may be alternative drugs for CN therapy.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Key Research and Development Program of China (grant no. 2016YFE0126000), China National Natural Science Foundation (grant no.81570392), China Postdoctoral Science Foundation funded project (grant no.2016M591937), Natural Science Fund for Colleges and Universities in Jiangsu Province (grant no.16KJB320017), and High Level Talent Support Program of Yangzhou University (grant no. 137080077).

Availability of data and materials

The data sets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JL and YW were major contributors in the conception and design of the research and revision of the manuscript. Acquisition of data was performed by WW, SG and LW were the major contributors in the analysis and interpretation of data and statistical analysis. Drafting the manuscript was performed by JL.

Ethics approval and consent to participate

Ethics Committee of Jiamusi University approved this study.

Patient consent for publication

Written informed consent was obtained from patient for publication of this research article and any accompanying images.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kim DG, Paek SH, Kim IH, Chi JG, Jung HW, Han DH, Choi KS and Cho BK: Central neurocytoma: The role of radiation therapy and long term outcome. Cancer. 79:1995–2002. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Schild SE, Scheithauer BW, Haddock MG, Schiff D, Burger PC, Wong WW and Lyons MK: Central neurocytomas. Cancer. 79:790–795. 1997. View Article : Google Scholar : PubMed/NCBI

3 

Schmidt MH, Gottfried ON, von Koch CS, Chang SM and McDermott MW: Central neurocytoma: A review. J Neurooncol. 66:377–384. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Kuo LM, Kuo CY, Lin CY, Hung MF, Shen JJ and Hwang TL: Intracellular glutathione depletion by oridonin leads to apoptosis in hepatic stellate cells. Molecules. 19:3327–3344. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Kadota S, Basnet P, Ishii E, Tamura T and Namba T: Antibacterial activity of trichorabdal A from Rabdosia trichocarpa against Helicobacter pylori. Zentralblatt fur Bakteriologie: Int J Med Microbiol. 286:63–67. 1997. View Article : Google Scholar

6 

Yang J, Jiang H, Wang C, Yang B, Zhao L, Hu D, Qiu G, Dong X and Xiao B: Oridonin triggers apoptosis in colorectal carcinoma cells and suppression of microRNA-32 expression augments oridonin-mediated apoptotic effects. Biomed Pharmacother. 72:125–134. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Wang H, Wang YF, Liu TG, Xiang XL and Huang SL: Experimental study on anti-pancreatic cancer effect of oridonin. Zhong Yao Cai. 37:1230–1233. 2014.(In Chinese). PubMed/NCBI

8 

Li Y, Wang Y, Wang S, Gao Y, Zhang X and Lu C: Oridonin phosphate-induced autophagy effectively enhances cell apoptosis of human breast cancer cells. Med Oncol. 32:3652015. View Article : Google Scholar : PubMed/NCBI

9 

Wang YY, Lv YF, Lu L and Cai L: Oridonin inhibits mTOR signaling and the growth of lung cancer tumors. Anticancer Drugs. 25:1192–1200. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Bohanon FJ, Wang X, Ding C, Ding Y, Radhakrishnan GL, Rastellini C, Zhou J and Radhakrishnan RS: Oridonin inhibits hepatic stellate cell proliferation and fibrogenesis. J Surg Res. 190:55–63. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Bu HQ, Liu DL, Wei WT, Chen L, Huang H, Li Y and Cui JH: Oridonin induces apoptosis in SW1990 pancreatic cancer cells via p53- and caspase-dependent induction of p38 MAPK. Oncol Rep. 31:975–982. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Zhang CL, Wu LJ, Tashiro S, Onodera S and Ikejima T: Oridonin induces a caspase-independent but mitochondria- and MAPKdependent cell death in the murine fibrosarcoma cell line L929. Biol Pharm Bull. 27:1527–1531. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Behrens J and Lustig B: The Wnt connection to tumorigenesis. Int J Dev Biol. 48:477–487. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Knoepfler PS, Cheng PF and Eisenman RN: N-myc is essential during neurogenesis for the rapid expansion of progenitor cell populations and the inhibition of neuronal differentiation. Genes Dev. 16:2699–2712. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Sim FJ, Keyoung HM, Goldman JE, Kim DK, Jung HW, Roy NS and Goldman SA: Neurocytoma is a tumor of adult neuronal progenitor cells. J Neurosci. 26:12544–12555. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Liu Y, Liu YZ, Zhang RX, Wang X, Meng ZJ, Huang J, Wu K, Luo JY, Zuo GW, Chen L, et al: Oridonin inhibits the proliferation of human osteosarcoma cells by suppressing Wnt/β-catenin signaling. Int J Oncol. 45:795–803. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Roy NS, Benraiss A, Wang S, Fraser RA, Goodman R, Couldwell WT, Nedergaard M, Kawaguchi A, Okano H and Goldman SA: Promoter-targeted selection and isolation of neural progenitor cells from the adult human ventricular zone. J Neurosci Res. 59:321–331. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Roy NS, Wang S, Jiang L, Kang J, Benraiss A, Harrison-Restelli C, Fraser RA, Couldwell WT, Kawaguchi A, Okano H, et al: In vitro neurogenesis by progenitor cells isolated from the adult human hippocampus. Nat Med. 6:271–277. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Lee SJ, Kim JE, Paek SH, Keyoung HM, Kim DG and Jung HW: Primary cell culture of central neurocytomas. J Korean Neurosurg Soc. 34:238–244. 2003.

20 

Wu K, Yang Q, Mu Y, Zhou L, Liu Y, Zhou Q and He B: Berberine inhibits the proliferation of colon cancer cells by inactivating Wnt/beta-catenin signaling. Int J Oncol. 41:292–298. 2012.PubMed/NCBI

21 

He TC, Zhou S, da Costa LT, Yu J, Kinzler KW and Vogelstein B: A simplified system for generating recombinant adenoviruses. Proc Natl Acad Sci USA. 95:2509–2514. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Tang N, Song WX, Luo J, Luo X, Chen J, Sharff KA, Bi Y, He BC, Huang JY, Zhu GH, et al: BMP-9-induced osteogenic differentiation of mesenchymal progenitors requires functional canonical Wnt/beta-catenin signalling. J Cell Mol Med. 13:2448–2464. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-delta delta C(t)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Feng M, Li J, Wang J, Ma C, Jiao Y, Wang Y, Zhang J, Sun Q, Ju Y, Gao L1 and Zhao Y: High glucose increases LPS-induced DC apoptosis through modulation of ERK1/2, AKT and Bax/Bcl-2. BMC Gastroenterol. 14:982014. View Article : Google Scholar : PubMed/NCBI

25 

Abelson PH: Medicine from plants. Science. 247:5131990. View Article : Google Scholar : PubMed/NCBI

26 

Liu H, Qian C and Shen Z: Anti-tumor activity of oridonin on SNU-5 subcutaneous xenograft model via regulation of c-Met pathway. Tumour Biol. 35:9139–9146. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Liu YQ, Mu ZQ, You S, Tashiro S, Onodera S and Ikejima T: Fas/FasL signaling allows extracelluar-signal regulated kinase to regulate cytochrome c release in oridonin-induced apoptotic U937 cells. Biol Pharm Bull. 29:1873–1879. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Jin S, Shen JN, Wang J, Huang G and Zhou JG: Oridonin induced apoptosis through Akt and MAPKs signaling pathways in human osteosarcoma cells. Cancer Biol Ther. 6:261–268. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Cheng Y, Qiu F, Ye YC, Tashiro S, Onodera S and Ikejima T: Oridonin induces G2/M arrest and apoptosis via activating ERK-p53 apoptotic pathway and inhibiting PTK-Ras-Raf-JNK survival pathway in murine fibrosarcoma L929 cells. Arch Biochem Biophys. 490:70–75. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Stennicke HR and Salvesen GS: Caspases-controlling intracellular signals by protease zymogen activation. Biochim Biophys Acta. 1477:299–306. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Gray DC, Mahrus S and Wells JA: Activation of specific apoptotic caspases with an engineered small molecule-activated protease. Cell. 142:637–646. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Clevers H: Wnt/beta-catenin signaling in development and disease. Cell. 127:469–480. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Wharton KA Jr: Runnin' with the Dvl: Proteins that associate with Dsh/Dvl and their significance to Wnt signal transduction. Dev Biol. 253:1–17. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Rao TP and Kühl M: An updated overview on Wnt signaling pathways: A prelude for more. Circ Res. 106:1798–1806. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Minde DP, Anvarian Z, Rüdiger SG and Maurice MM: Messing up disorder: How do missense mutations in the tumor suppressor protein APC lead to cancer? Mol Cancer. 10:1012011. View Article : Google Scholar : PubMed/NCBI

36 

Minde DP, Radli M, Forneris F, Maurice MM and Stefan Rüdiger GD: large extent of disorder in adenomatous polyposis coli offers a strategy to guard Wnt signalling against point mutations. PLoS One. 8:e772572013. View Article : Google Scholar : PubMed/NCBI

37 

MacDonald BT, Tamai K and He X: Wnt/beta-catenin signaling: Components, mechanisms, and diseases. Dev Cell. 17:9–26. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Lopez J and Tait SW: Mitochondrial apoptosis: Killing cancer using the enemy within. Br J Cancer. 112:957–962. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Cohen GM: Caspases: The executioners of apoptosis. Biochem J. 326:1–16. 1997. View Article : Google Scholar : PubMed/NCBI

40 

Tetsu O and McCormick F: Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature. 398:422–426. 1999. View Article : Google Scholar : PubMed/NCBI

41 

Reya T and Clevers H: Wnt signalling in stem cells and cancer. Nature. 434:843–850. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Damalas A, Ben-Ze'ev A, Simcha I, Shtutman M, Leal JF, Zhurinsky J, Geiger B and Oren M: Excess beta-catenin promotes accumulation of transcriptionally active p53. EMBO J. 18:3054–3063. 1999. View Article : Google Scholar : PubMed/NCBI

43 

He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B and Kinzler KW: Identification of c-MYC as a target of the APC pathway. Science. 281:1509–1512. 1998. View Article : Google Scholar : PubMed/NCBI

44 

Klaus A and Birchmeier W: Wnt signalling and its impact on development and cancer. Nat Rev Cancer. 8:387–398. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Li CY, Wang EQ, Cheng Y and Bao JK: Oridonin: An active diterpenoid targeting cell cycle arrest, apoptotic and autophagic pathways for cancer therapeutics. Int J Biochem Cell Biol. 43:701–704. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Li X, Li X, Wang J, Ye Z and Li JC: Oridonin up-regulates expression of P21 and induces autophagy and apoptosis in human prostate cancer cells. Int J Biol Sci. 8:901–912. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Hu HZ, Yang YB and Xu XD: Oridonin induces apoptosis via PI3K/Akt pathway in cervical carcinoma HeLa cell line. Acta Pharmacol Sin. 28:1819–1826. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 16 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liang J, Wang W, Wei L, Gao S and Wang Y: Oridonin inhibits growth and induces apoptosis of human neurocytoma cells via the Wnt/β‑catenin pathway. Oncol Lett 16: 3333-3340, 2018
APA
Liang, J., Wang, W., Wei, L., Gao, S., & Wang, Y. (2018). Oridonin inhibits growth and induces apoptosis of human neurocytoma cells via the Wnt/β‑catenin pathway. Oncology Letters, 16, 3333-3340. https://doi.org/10.3892/ol.2018.8977
MLA
Liang, J., Wang, W., Wei, L., Gao, S., Wang, Y."Oridonin inhibits growth and induces apoptosis of human neurocytoma cells via the Wnt/β‑catenin pathway". Oncology Letters 16.3 (2018): 3333-3340.
Chicago
Liang, J., Wang, W., Wei, L., Gao, S., Wang, Y."Oridonin inhibits growth and induces apoptosis of human neurocytoma cells via the Wnt/β‑catenin pathway". Oncology Letters 16, no. 3 (2018): 3333-3340. https://doi.org/10.3892/ol.2018.8977