Gambogenic acid induces proteasomal degradation of CIP2A and sensitizes hepatocellular carcinoma to anticancer agents

  • Authors:
    • Xian‑Jun Yu
    • Qun Zhao
    • Xuan‑Bin Wang
    • Jing‑Xuan Zhang
    • Xiao‑Bo Wang
  • View Affiliations

  • Published online on: October 20, 2016     https://doi.org/10.3892/or.2016.5188
  • Pages: 3611-3618
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncoprotein that is overexpressed in many human malignancies. It regulates phosphorylated AKT and stabilizes c‑Myc in cell proliferation and tumor formation, suggesting that CIP2A plays an essential role in the development of cancer. In the present study, we report that a natural compound, gambogenic acid (GEA), induced the degradation of CIP2A via the ubiquitin‑proteasome pathway. Interestingly, the combination of GEA and proteasome inhibitors potentiated the accumulation of ubiquitinated CIP2A and aggresome formation. In addition, GEA exhibited an inhibitory effect on cell proliferation and CIP2A‑downstream signaling molecules (c‑Myc and pAKT). Furthermore, GEA and CIP2A silencing enhanced the chemosensitivity of hepatocellular carcinoma cells to anticancer agents, suggesting that a combination of a CIP2A inhibitor and anticancer agents could be a valuable clinical therapeutic strategy. These results indicate that GEA is a CIP2A inhibitor that interferes with the ubiquitination and destabilization of CIP2A, providing a promising strategy to enhance the combinational therapy for hepatocellular carcinoma.

Introduction

Human hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide and ranks third in terms of global cancer-related mortality (1). Liver cirrhosis is believed to be the most significant risk factor for 70–90% of HCC patients (2). Moreover, viral infection, liver cytotoxicity, chronic inflammation, and many other factors have been implicated in HCC progression (3). Unfortunately, although some improvement has been achieved in the clinic, overall the prognosis is poor due to the development of resistance to chemotherapy and radiotherapy (4). Therefore, the development of novel targeted anticancer agents is extremely important to overcome this disease.

Cancerous inhibitor of protein phosphatase 2A (CIP2A) stabilizes the c-Myc protein via suppressing the protein phosphatase 2A (PP2A) (5). It has been reported to be amplified or overexpressed in a wide variety of human malignancies, such as gastric (6), breast (7), renal cell (8), bladder (9), and lung (10) cancer. The functional roles of CIP2A involve cell growth, cell invasion, drug resistance, and tumor formation (1012). Several compounds from traditional Chinese medicine have been reported to exhibit anticancer activity via degradation of CIP2A and subsequent inactivation of AKT (13,14). These findings indicate that CIP2A could be a promising target for cancer chemotherapy.

The genus Garcinia is known for its rich variety of oxygenated and prenylated phenol derivatives. Gambogic acid is a major active component of gamboge isolated from the resin of Garcinia hanburyi, which has been shown to have potent anticancer activity and is authorized to be tested in clinical trails (1518). Gambogenic acid (GEA) is another active component of gamboge which exhibits cytotoxicity and anti-inflammatory activity (19,20). The molecular mechanisms that underlie the effects of GEA include induction of cell cycle arrest (21,22), apoptosis 21,2327, autophagy (22,28), necroptosis (29) and chemosensitivity (29,30). Herein, we demonstrated that GEA induced rapid proteasome-mediated degradation of CIP2A. GEA also showed potent anticancer activity and enhanced the effect of chemotherapeutic agents against HCC.

Materials and methods

Chemicals and reagents

GEA was extracted from gamboges by Dr Quanbin Han as previously described (20), dissolved in dimethyl sulfoxide (DMSO; Sigma-Aldrich, St. Louis, MO, USA) to make a stock solution (20 mM) and stored at −20°C. NH4Cl and 3-MA were purchased from Sigma-Aldrich. Cycloheximide (CHX) and nocodazole were obtained from Beyotime Institute of Biotechnology (Jiangsu, China). CellTiter 96 AQueous One Solution Cell Proliferation Assay and Z-VAD-FMK were obtained from Promega (Madison, WI, USA). Antibodies used in our study were as follows: anti-β-actin (Sigma-Aldrich); anti-CIP2A, c-Myc, ubiquitin (Santa Cruz Biotechnology); anti-pAKT (Ser473) (Cell Signaling Technology); and anti-rabbit and anti-mouse HRP-conjugated secondary antibodies (Pierce). Detection was performed using a Chemiluminescent Western Blot Detection kit (Thermo Fisher Scientific, Rockford, IL, USA).

Cell culture

The human hepatoma cell lines Hep G2 and Bel-7402 were obtained from the American Tissue Culture Collection (ATCC; Manassas, VA, USA) and maintained in Dulbecco's modified Eagle's medium (DMEM; Gibco-BRL, Gaithersburg, MD, USA) supplemented with 10% fetal bovine serum (FBS; Biological Industries, Kibbutz Beit-Haemek, Israel), 100 U/ml penicillin, 100 µg/ml streptomycin, and 2 mM glutamine (Invitrogen Life Technologies, Carlsbad, CA, USA). All cells were cultured in a humidified incubator at 37°C under 5% CO2.

Cell viability assay

Cells were seeded in 96-well plates (1×103 cells/well) and then exposed to the indicated agents. After incubation for the indicated time, cell viability assay was conducted using the cell titer assay. Cell growth curve was estimated using trypan blue dye exclusion.

Real-time reverse transcription-polymerase chain reaction (RT-PCR)

Total RNA was isolated from cells by using the TRIzol reagent (Invitrogen Life Technologies) according to the manufacturer's instructions. The first strand complementary DNA (cDNA) was synthesized using PrimeScript™ RT reagent kit with gDNA Eraser (Takara, Dalian, China). Primers used for RT-PCR analysis of human CIP2A included sense, 5′-CCATATGCTCACTCAGATGATGT-3′ and antisense, 5′-GTGTATCATCTCCACAGAGAGTT-3′; and for GAPDH sense, 5′-TCACCAGGGCTGCTTTTA-3′ and antisense, 5′-AAGGTCATCCCTGAGCTGAA-3′. PCR products were separated on 1.5% agarose gels and stained with GoldView. To confirm CIP2A mRNA expression, real-time polymerase chain reaction (qRT-PCR) was performed using the SYBR Premix Ex Taq (Takara). The primers were as follows: CIP2A sense, 5′-TGCGGCACTTGGAGGTAATTTC-3′ and antisense, 5′-AGCTCTACAAGGCAACTCAAGC-3′; actin sense, 5′-ATCGTCCACCGCAAATGCTTCTA-3′ and antisense, 5′-AGCCATGCCAATCTCATCTTGTT-3′. The amplifications were performed as follows: 94°C for 10 min and then 40 cycles of 94°C for 15 sec, 60°C for 30 sec and 72°C for 30 sec. Quantified values for gene expression were generated by the relative quantification method.

siRNA assays

Cells were transfected with double-stranded siRNA oligonucleotides (100 nM) in 6-well plates using Lipofectamine 2000 (Invitrogen Life Technologies) according to the manufacturer's instructions. The sequences of siRNAs are as follows: CIP2A siRNA, 5′-GGUGCACGUUUCAUCAAUU-3′; NC siRNA, 5′-GGUGCACGUUUCAUCAAUU-3′.

Western blotting

Cells were suspended in lysis buffer containing 50 mM Tris-HCl (pH 7.6), 150 mM NaCl, 1% NP40, 1 mM EDTA, 1 mM Na3VO4, 1 mM NaF, and a cocktail of 1 mM PMSF and 1 mM protease inhibitors. The lysates were centrifuged at 12,000 × g for 10 min at 4°C, followed by measurements of protein concentrations using Pierce BCA Protein Assay Kit (Thermo Fisher Scientific). The supernatants were collected as NP40-soluble fractions. The pellets (NP40-insoluble fractions) were lysed in lysis buffer containing 2% SDS and boiled at 100°C for 7 min and chilled on ice. Proteins (20 µg) were separated on 10% SDS-PAGE gel and transferred to PVDF membranes (Millipore Corp., Billerica, MA, USA). The membranes were incubated overnight with specific primary antibodies at 4°C after being blocked with 5% non-fat milk. After being washed three times with PBS containing 0.05% Tween-20 (PBST), the membranes were incubated with HRP-conjugated secondary antibodies for 1 h at room temperature, followed by 3-times washing with 0.05% Tween-20/PBS and then detected using chemiluminescent substrate.

Statistical analysis

Quantitative data are presented as means ± SED from triplicate experiments. Comparison between groups was performed by ANOVA and P≤0.05 was considered to indicate a statistically significant difference.

Results

GEA triggers degradation of CIP2A

We aimed to ascertain whether GEA (Fig. 1A) affects CIP2A protein expression. We demonstrated that following treatment with GEA at 2.5 µM for 2–12 h, a dramatic decrease in CIP2A expression in Hep G2 cells was observed (Fig. 1B). Similarly, treatment with GEA at 1.0–2.5 µM for 12 h decreased the CIP2A expression in a dose-dependent manner (Fig. 1B). These observations were further confirmed in Bel-7402 cells (Fig. 1C). Treatment with GEA at 2.5 µM for 6–12 h also suppressed CIP2A in other hepatocellular carcinomas (Hep 3B, HuH-7), lung (A549), breast (MCF-7) and colon (HT-29) cancer cell lines, indicating that GEA-induced CIP2A downregulation is not cell-type specific (Fig. 1D).

GEA downregulates CIP2A at the post-transcriptional level

In order to clarify the underlying mechanism involved in CIP2A downregulation, RT-PCR assays were performed and revealed that GEA at 2.5 µM for 6 h did not exhibit inhibitory effects on the expression of CIP2A mRNA in Hep G2 cells (Fig. 2A). These similar observations were confirmed by quantitative RT-PCR (Fig. 2B). Since GEA-mediated downregulation of CIP2A is not associated with transcription, we hypothesized that the reduction in CIP2A might be due to protein stabilization and degradation. As shown in Fig. 2C and D, protein synthesis inhibitor CHX barely reduced the expression of CIP2A within 8 h, however, the combination of GEA and CHX resulted in a marked reduction of CIP2A at the protein level within 4 h. These results indicate that GEA decreased CIP2A at the post-transcription level.

GEA triggers ubiquitin-proteasome-mediated degradation of CIP2A

Four major proteolytic systems mediate protein stability: caspase, calpain, lysosome and proteasome (31). The caspase family of cysteine proteases is involved in cell death and cleavage of substrate proteins (32). Hep G2 cells were pre-treated with pan-caspase inhibitor Z-VAD-fmk (Z-VAD) for 2 h, followed by treatment with or without GEA for 6 h. However, no significant reversal effect was observed in the presence of Z-VAD (Fig. 3A). Calpains represent a well-conserved family of calcium-dependent cysteine proteases (33). We then pre-treated Hep G2 cells with calpain inhibitor III MDL-28170 (MDL), and the degradation of CIP2A was not reversed (Fig. 3B). Lysosomes and autophagosomes are organelles which play a central role in the control of cell fate (34). The Hep G2 cells were pretreated with a lysosomal protease inhibitor (NH4Cl) and an autophagy inhibitor (3-MA) and then treated with GEA. The two inhibitors did not prevent GEA-induced CIP2A degradation (Fig. 3C and D). The ubiquitin-proteasome pathway plays an important role in intracellular proteolysis (35,36). Interestingly, we found that following treatment with the proteasome inhibitor MG132 or PSI alone, the expression of CIP2A in the Hep G2 cells was not affected within 6 h (Fig. 3E). However, the combination of MG132/PSI and GEA markedly impaired CIP2A degradation (Fig. 3E). Previous studies have reported that the proteasome inhibitor promotes accumulation of ubiquitinated proteins and shifts them into detergent-insoluble cellular fractions, suggestive for aggresomes (37). To determine the distribution of CIP2A, Hep G2 cells were pre-treated with MG132 followed by GEA treatment. The results showed that accumulation of CIP2A in NP40 was induced in the insoluble fractions, while it had minimal effect on GEA-treated controls (Fig. 3F). Similar results were noted with GEA in combination with PSI in the Hep G2 cells (Fig. 3F). To further elucidate the molecular mechanism underlying the above process, we immunoprecipitated CIP2A from the NP40-insoluble fraction after treatment with GEA alone, MG132 alone, or their combinations. Notably, GEA alone was able to accumulate the ubiquitinated CIP2A (Fig. 3G, lane 3). Most importantly, these effects were enhanced following treatment with the combination of MG132 and GEA (Fig. 3G, lane 5). Aggresome formation is related to redistribution of the intermediate filament protein and blocked by microtubule depolymerizing (38,39). We showed that microtubule depolymerizing agent nocodazole prevented the levels of CIP2A in the NP40 insoluble fraction (Fig. 3H, lane 6 vs. 4). Expectedly, an increase in CIP2A levels was detected in the NP40-soluble fraction (Fig. 3H, lane 6 vs. 4). These results suggest that GEA stimulates ubiquitin proteasome-mediated degradation of CIP2A.

Figure 3.

GEA triggers ubiquitin proteasome-mediated degradation of CIP2A. (A-D) Hep G2 cells were pre-treated with (A) Z-VAD (20 µM), (B) MDL (40 µM), (C) NH4Cl (20 mM), (D) 3-MA (10 mM) for 2 h, followed by GEA treatment (2.5 µM) for 6 h and the protein level of CIP2A was detected. (E) Hep G2 cells were pre-treated with MG132 (10 µM) or PSI (10 µM) for 2 h, followed by GEA treatment (2.5 µM) for 6 h. Cell lysates were subjected to western blotting using the anti-CIP2A antibody. (F) Hep G2 cells were treated with MG132 (10 µM) or PSI (10 µM) for 2 h prior to GEA treatment for 6 h. Cells were harvested and lysed in non-NP40 lysis buffer. The soluble and insoluble fractions were separated, boiled in SDS buffer for 10 min, and analyzed. Protein expression was quantified by densitometric analysis and normalized against β-actin expression. (G) Effect of GEA on CIP2A ubiquitination. Hep G2 cells were pre-treated with MG132 (10 µM) for 2 h, followed by GEA treatment for 1 h. NP40-insoluble fractions were extracted as described above and subjected to immunoprecipitation with the CIP2A antibody, followed by western blotting using ubiquitin antibody against ubiquitinated CIP2A. (H) Effect of GEA on aggresome formation in the presence of proteasome inhibitor MG132. Hep G2 cells were pre-treated with MG132 (10 µM) and/or nocodazole (20 µg/ml) for 2 h, followed by GEA incubation (2.5 µM) for 6 h. The soluble and insoluble fractions were separated in cell lysis, and then analyzed by western blotting. Protein expression was quantified by densitometric analysis and normalized against β-actin expression. GEA, gambogenic acid; CIP2A, cancerous inhibitor of protein phosphatase 2A. Data are presented as means ± SEM. **P<0.01, ***P<0.001.

GEA suppresses cell proliferation and downregulates CIP2A-downstream molecules

CIP2A is a candidate therapeutic target and inhibition of its activity has potent anticancer effects (40). We therefore tested the effects of GEA on HCC cells, and found that GEA inhibited cell proliferation in a dose- and time-dependent manner (Fig. 4A and B). To determine whether GEA is more sensitive to tumor than normal cells, we examined the effect of GEA on normal human hepatocyte HL-7702 and mouse primary hepatocyte cells. We found that its cytotoxic effects on normal liver cells was weak (Fig. 4C), suggesting that GEA selectively affects tumor cells. Previous studies have demonstrated that CIP2A may activate AKT and modulate c-Myc stability (41). We therefore detected the effects of GEA on c-Myc and pAKT. We found that GEA markedly dowregulated c-Myc and pAKT in the Hep G2 and Bel-7402 cell lines (Fig. 4D). These results indicate that CIP2A-pAKT may play an important role in the inhibitory effect of GEA against tumor cells.

GEA and CIP2A silencing enhance the sensitivity to chemotherapeutic agents

Previous studies demonstrated that CIP2A overexpression is associated with drug resistance (12,42). AKT is a key mediator of cell survival and resistance to chemotherapy (43). We then proceeded to evaluate whether GEA enhances chemosensitivity. Our data showed that pre-treatment with GEA increased the cytotoxic effects compared to treatment with Taxol, cisplatin (CDDP) and 5-fluorouracil (5-FU) alone, respectively (Fig. 5A). We next explored the role of CIP2A in GEA-induced sensitivity in HCC. We showed that knockdown of CIP2A also enhanced the sensitivity of HCC to chemotherapy agents (Fig. 5B). These results imply that GEA sensitizes HCC to chemotherapy, and this effect is associated with CIP2A expression.

Discussion

CIP2A is strongly implicated in carcinogenesis which is associated with cigarette smoking and Helicobacter pylori infection, suggesting that CIP2A can serve as an indicator for assessing the potential carcinogenic risk (10,44). Furthermore, overexpression of CIP2A is associated with drug resistance and tumor formation. Moreover, overexpression of CIP2A is frequently noted in most human cancers, and implies an indicator of poor clinical outcomes (5,12). CIP2A-targeting compounds such as bortezomib (12), ethoxysanguinarine (13), and celastrol (14) exert chemopreventive effects. Herein, we report that GEA is a new CIP2A inhibitor which enhances the chemosensitivity of HCC to chemotherapeutic agents. We further showed that GEA triggered degradation of CIP2A in HCC, lung, breast and colon cancer cell lines (Fig. 1). Since CIP2A can be regulated at the transcriptional and post-transcription levels, subsequent data demonstrated that GEA did not inhibit CIP2A mRNA, but decreased the half-life of CIP2A protein (Fig. 2). These results suggest that GEA induced proteolytic degradation of CIP2A.

We further explored the mechanisms underlying the GEA-induced CIP2A degradation. Protein instability and degradation, such as caspase or calpain cleavage, lysosomal or autophagic protein degradation, and proteasome-mediated degradation, play a critical role in proteolysis (31). We focused on these and found that proteasome inhibitors impaired the GEA-induced CIP2A degradation, while inhibitors of caspases, calpain, lysosomes and autophagosomes had no effect on GEA-induced CIP2A degradation (Fig. 3A-E). Emerging evidence previously showed that proteasome inhibitors promote the accumulation of ubiquitinated CIP2A and aggresome formation (14). We observe herein that the proteasome inhibitors were able to protect CIP2A from GEA-mediated degradation (Fig. 3F). This phenomenon may be associated with accumulation of insoluble ubiquitinated CIP2A, leading to their aggregation and deposition in detergent-insoluble fractions (12). Indeed, treatment with GEA or MG132 induced accumulation of ubiquitinated CIP2A (Fig. 3G). Importantly, a significant increase was observed following co-treatment with GEA and MG132 (Fig. 3G). These results further confirmed that GEA is a proteasome inhibitor and GEA treatment promotes CIP2A ubiquitination and accumulation. Previous evidence revealed that aggresome formation is associated with microtubule disruption (38). Consistent with this, our study found that microtubule depolymerizing agent nocodazole suppressed CIP2A aggregation in NP40-insoluble fractions (Fig. 3H). In addition, the levels of CIP2A were significantly enhanced in the NP40-soluble fractions, suggesting that aggresome formation is an important process in GEA-triggered degradation of CIP2A (Fig. 3H). Collectively, we speculate that proteasome inhibition may cause protein misfolding and aggregation, leading to accumulation of ubiquitinated CIP2A in NP40-insoluble fractions. These findings partially indicate that GEA-induced CIP2A degradation is mediated by the ubiquitin-proteasome pathway, but the detailed mechanisms, including the binding type (directly or indirectly), sites and the E3 ligase remain unclear and warrant further investigation.

Several studies have shown that GEA has a potent anticancer effect in various cancer cells in vitro and in vivo. However, the mechanisms underlying the anticancer activity remain unclear. Herein, we showed that GEA suppressed the proliferation and cell growth in HCC cell lines, but had a mild effect on normal liver cells (Fig. 4A-C). Interestingly, the expression of CIP2A-downstream molecules c-Myc and pAKT were impaired in cells following GEA treatment (Fig. 4D). These findings suggest that GEA potentiates the inhibitory effect via affecting the CIP2A-pAKT signaling pathway. Recent evidence suggests that bortezomib serves as a CIP2A inhibitor and enhances the effect of radiotherapy dependent on the CIP2A-PP2A-AKT signaling network, indicating that CIP2A may be associated with drug resistance (12). GEA was found to synergize with the cytotoxicity of chemotherapeutic drugs (Fig. 5A). Importantly, CIP2A-silenced cells were also more sensitive to chemotherapeutic agents (Fig. 5B). These results indicate that GEA as a CIP2A-targeting inhibitor modulates CIP2A and enhances the sensitivity of HCC cells to multiple chemotherapeutic agents.

In summary, we provide initial evidence that GEA triggered the degradation of CIP2A in various cancer cell lines. This rapid degradation may be associated with the ubiquitin-proteasome pathway. In addition, GEA suppressed the CIP2A-pAKT pathway and enhanced sensitivity to chemotherapeutic agents. Our findings indicate that GEA is a novel CIP2A inhibitor that may have therapeutic potential in HCC. Admittedly, the detailed mechanisms of GEA-induced CIP2A degradation remain unclear and warrant further investigation.

Acknowledgements

We thank Dr Haibing Zhang and Dr Quanbin Han for the long-term support. This study was supported by grants from the National Natural Science Foundation of China (81502548), the Natural Science Foundation of Hubei Province of China (2015CFB210), the Natural Science Foundation of Hubei Provincial Department of Education (Q20152101), the Young Scientist Innovation Team Project of Hubei Colleges (T201510), the Scientific and Technological Project of Shiyan City of Hubei Province (15Y08), the Initial Project for Post-Graduates of Hubei University of Medicine (2014QDJZR09), the Foundation for Innovative Research Team of Hubei University of Medicine (2014 CXX02) and Thousand Young Talents Program of the Chinese government.

References

1 

Wang X, Zhang A and Sun H: Power of metabolomics in diagnosis and biomarker discovery of hepatocellular carcinoma. Hepatology. 57:2072–2077. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Fattovich G, Giustina G, Christensen E, Pantalena M, Zagni I, Realdi G and Schalm SW: Influence of hepatitis delta virus infection on morbidity and mortality in compensated cirrhosis type B. The European Concerted Action on Viral Hepatitis (Eurohep). Gut. 46:420–426. 2000. View Article : Google Scholar : PubMed/NCBI

3 

El Tayebi HM, Omar K, Hegy S, El Maghrabi M, El Brolosy M, Hosny KA, Esmat G and Abdelaziz AI: Repression of miR-17-5p with elevated expression of E2F-1 and c-MYC in non-metastatic hepatocellular carcinoma and enhancement of cell growth upon reversing this expression pattern. Biochem Biophys Res Commun. 434:421–427. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Fujino H, Kimura T, Aikata H, Miyaki D, Kawaoka T, Kan H, Fukuhara T, Kobayashi T, Naeshiro N, Honda Y, et al: Role of 3-D conformal radiotherapy for major portal vein tumor thrombosis combined with hepatic arterial infusion chemotherapy for advanced hepatocellular carcinoma. Hepatol Res. 45:607–617. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Junttila MR, Puustinen P, Niemelä M, Ahola R, Arnold H, Böttzauw T, Ala-aho R, Nielsen C, Ivaska J, Taya Y, et al: CIP2A inhibits PP2A in human malignancies. Cell. 130:51–62. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Li W, Ge Z, Liu C, Liu Z, Björkholm M, Jia J and Xu D: CIP2A is overexpressed in gastric cancer and its depletion leads to impaired clonogenicity, senescence, or differentiation of tumor cells. Clin Cancer Res. 14:3722–3728. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Côme C, Laine A, Chanrion M, Edgren H, Mattila E, Liu X, Jonkers J, Ivaska J, Isola J, Darbon JM, et al: CIP2A is associated with human breast cancer aggressivity. Clin Cancer Res. 15:5092–5100. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Basile JR and Czerninski R: The role of CIP2A in oral squamous cell carcinoma. Cancer Biol Ther. 10:700–702. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Huang LP, Savoly D, Sidi AA, Adelson ME, Mordechai E and Trama JP: CIP2A protein expression in high-grade, high-stage bladder cancer. Cancer Med. 1:76–81. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Ma L, Wen ZS, Liu Z, Hu Z, Ma J, Chen XQ, Liu YQ, Pu JX, Xiao WL, Sun HD, et al: Overexpression and small molecule-triggered downregulation of CIP2A in lung cancer. PLoS One. 6:e201592011. View Article : Google Scholar : PubMed/NCBI

11 

Ren J, Li W, Yan L, Jiao W, Tian S, Li D, Tang Y, Gu G, Liu H and Xu Z: Expression of CIP2A in renal cell carcinomas correlates with tumour invasion, metastasis and patients' survival. Br J Cancer. 105:1905–1911. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Huang CY, Wei CC, Chen KC, Chen HJ, Cheng AL and Chen KF: Bortezomib enhances radiation-induced apoptosis in solid tumors by inhibiting CIP2A. Cancer Lett. 317:9–15. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Liu Z, Ma L, Wen ZS, Cheng YX and Zhou GB: Ethoxysanguinarine induces inhibitory effects and downregulates CIP2A in lung cancer cells. ACS Med Chem Lett. 5:113–118. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Liu Z, Ma L, Wen ZS, Hu Z, Wu FQ, Li W, Liu J and Zhou GB: Cancerous inhibitor of PP2A is targeted by natural compound celastrol for degradation in non-small-cell lung cancer. Carcinogenesis. 35:905–914. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Pandey MK, Sung B, Ahn KS, Kunnumakkara AB, Chaturvedi MM and Aggarwal BB: Gambogic acid, a novel ligand for transferrin receptor, potentiates TNF-induced apoptosis through modulation of the nuclear factor-kappaB signaling pathway. Blood. 110:3517–3525. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Wang X, Chen Y, Han QB, Chan CY, Wang H, Liu Z, Cheng CH, Yew DT, Lin MC, He ML, et al: Proteomic identification of molecular targets of gambogic acid: Role of stathmin in hepatocellular carcinoma. Proteomics. 9:242–253. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Wang J, Chi Y, Zhan XK, Xie GR, Wang ZZ, Xiao W, Wang YG, Hu JF, Yu H, Yang L, et al: An open-labeled, randomized, multicentered, phase IIa study for advanced cancer treatment by gambogic acid injection (THS). J Clin Oncol. 29:e130952011.

18 

Chi Y, Wang J, Zhan X, Xie G, Wang Z, Xiao W, Wang Y, Hu J, Yu H, Yang L, et al: p53 open-label, randomised, multicentre, phase 2a study of gambogic acid injection (THS) for treatment of advanced cancer. EJC Suppl. 9:212011. View Article : Google Scholar

19 

Yu X, Zhao Q, Zhang H, Fan C, Zhang X, Xie Q, Xu C, Liu Y, Wu X, Han Q, et al: Gambogenic acid inhibits LPS-simulated inflammatory response by suppressing NF-κB and MAPK in macrophages. Acta Biochim Biophys Sin (Shanghai). 48:454–461. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Han QB, Wang YL, Yang L, Tso TF, Qiao CF, Song JZ, Xu LJ, Chen SL, Yang DJ and Xu HX: Cytotoxic polyprenylated xanthones from the resin of Garcinia hanburyi. Chem Pharm Bull (Tokyo). 54:265–267. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Li Q, Cheng H, Zhu G, Yang L, Zhou A, Wang X, Fang N, Xia L, Su J, Wang M, et al: Gambogenic acid inhibits proliferation of A549 cells through apoptosis-inducing and cell cycle arresting. Biol Pharm Bull. 33:415–420. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Yu XJ, Han QB, Wen ZS, Ma L, Gao J and Zhou GB: Gambogenic acid induces G1 arrest via GSK3β-dependent cyclin D1 degradation and triggers autophagy in lung cancer cells. Cancer Lett. 322:185–194. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Cheng H, Su JJ, Peng JY, Wang M, Wang XC, Yan FG, Wang XS and Li QL: Gambogenic acid inhibits proliferation of A549 cells through apoptosis inducing through up-regulation of the p38 MAPK cascade. J Asian Nat Prod Res. 13:993–1002. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Yan F, Wang M, Chen H, Su J, Wang X, Wang F, Xia L and Li Q: Gambogenic acid mediated apoptosis through the mitochondrial oxidative stress and inactivation of Akt signaling pathway in human nasopharyngeal carcinoma CNE-1 cells. Eur J Pharmacol. 652:23–32. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Chen HB, Zhou LZ, Mei L, Shi XJ, Wang XS, Li QL and Huang L: Gambogenic acid-induced time- and dose-dependent growth inhibition and apoptosis involving Akt pathway inactivation in U251 glioblastoma cells. J Nat Med. 66:62–69. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Yan F, Wang M, Li J, Cheng H, Su J, Wang X, Wu H, Xia L, Li X, Chang HC, et al: Gambogenic acid induced mitochondrial-dependent apoptosis and referred to phospho-Erk1/2 and phospho-p38 MAPK in human hepatoma HepG2 cells. Environ Toxicol Pharmacol. 33:181–190. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Zhou J, Luo YH, Wang JR, Lu BB, Wang KM and Tian Y: Gambogenic acid induction of apoptosis in a breast cancer cell line. Asian Pac J Cancer Prev. 14:7601–7605. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Mei W, Dong C, Hui C, Bin L, Fenggen Y, Jingjing S, Cheng P, Meiling S, Yawen H, Xiaoshan W, et al: Gambogenic acid kills lung cancer cells through aberrant autophagy. PLoS One. 9:e836042014. View Article : Google Scholar : PubMed/NCBI

29 

Su J, Cheng H, Zhang D, Wang M, Xie C, Hu Y, Chang HC and Li Q: Synergistic effects of 5-fluorouracil and gambogenic acid on A549 cells: Activation of cell death caused by apoptotic and necroptotic mechanisms via the ROS-mitochondria pathway. Biol Pharm Bull. 37:1259–1268. 2014. View Article : Google Scholar : PubMed/NCBI

30 

He Y, Ding J, Lin Y, Li J, Shi Y, Wang J, Zhu Y, Wang K and Hu X: Gambogenic acid alters chemosensitivity of breast cancer cells to Adriamycin. BMC Complement Altern Med. 15:1812015. View Article : Google Scholar : PubMed/NCBI

31 

Wang Y and Zhang Y: Regulation of TET protein stability by calpains. Cell Reports. 6:278–284. 2014. View Article : Google Scholar : PubMed/NCBI

32 

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

33 

Storr SJ, Carragher NO, Frame MC, Parr T and Martin SG: The calpain system and cancer. Nat Rev Cancer. 11:364–374. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Medina DL, Di Paola S, Peluso I, Armani A, De Stefani D, Venditti R, Montefusco S, Scotto-Rosato A, Prezioso C, Forrester A, et al: Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat Cell Biol. 17:288–299. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Shen M, Schmitt S, Buac D and Dou QP: Targeting the ubiquitin-proteasome system for cancer therapy. Expert Opin Ther Targets. 17:1091–1108. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Ciechanover A: Proteolysis: From the lysosome to ubiquitin and the proteasome. Nat Rev Mol Cell Biol. 6:79–87. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Wang J, Zhao Q, Qi Q, Gu HY, Rong JJ, Mu R, Zou MJ, Tao L, You QD and Guo QL: Gambogic acid-induced degradation of mutant p53 is mediated by proteasome and related to CHIP. J Cell Biochem. 112:509–519. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Johnston JA, Ward CL and Kopito RR: Aggresomes: A cellular response to misfolded proteins. J Cell Biol. 143:1883–1898. 1998. View Article : Google Scholar : PubMed/NCBI

39 

Bence NF, Sampat RM and Kopito RR: Impairment of the ubiquitin-proteasome system by protein aggregation. Science. 292:1552–1555. 2001. View Article : Google Scholar : PubMed/NCBI

40 

Khanna A, Rane JK, Kivinummi KK, Urbanucci A, Helenius MA, Tolonen TT, Saramäki OR, Latonen L, Manni V, Pimanda JE, et al: CIP2A is a candidate therapeutic target in clinically challenging prostate cancer cell populations. Oncotarget. 6:19661–19670. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Khanna A, Böckelman C, Hemmes A, Junttila MR, Wiksten JP, Lundin M, Junnila S, Murphy DJ, Evan GI, Haglund C, et al: MYC-dependent regulation and prognostic role of CIP2A in gastric cancer. J Natl Cancer Inst. 101:793–805. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Choi YA, Park JS, Park MY, Oh KS, Lee MS, Lim JS, Kim KI, Kim KY, Kwon J, Yoon DY, et al: Increase in CIP2A expression is associated with doxorubicin resistance. FEBS Lett. 585:755–760. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Jiao M and Nan KJ: Activation of PI3 kinase/Akt/HIF-1α pathway contributes to hypoxia-induced epithelial-mesenchymal transition and chemoresistance in hepatocellular carcinoma. Int J Oncol. 40:461–468. 2012.PubMed/NCBI

44 

Zhao D, Liu Z, Ding J, Li W, Sun Y, Yu H, Zhou Y, Zeng J, Chen C and Jia J: Helicobacter pylori CagA upregulation of CIP2A is dependent on the Src and MEK/ERK pathways. J Med Microbiol. 59:259–265. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 36 Issue 6

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
Yu XJ, Zhao Q, Wang XB, Zhang JX and Wang XB: Gambogenic acid induces proteasomal degradation of CIP2A and sensitizes hepatocellular carcinoma to anticancer agents. Oncol Rep 36: 3611-3618, 2016
APA
Yu, X., Zhao, Q., Wang, X., Zhang, J., & Wang, X. (2016). Gambogenic acid induces proteasomal degradation of CIP2A and sensitizes hepatocellular carcinoma to anticancer agents. Oncology Reports, 36, 3611-3618. https://doi.org/10.3892/or.2016.5188
MLA
Yu, X., Zhao, Q., Wang, X., Zhang, J., Wang, X."Gambogenic acid induces proteasomal degradation of CIP2A and sensitizes hepatocellular carcinoma to anticancer agents". Oncology Reports 36.6 (2016): 3611-3618.
Chicago
Yu, X., Zhao, Q., Wang, X., Zhang, J., Wang, X."Gambogenic acid induces proteasomal degradation of CIP2A and sensitizes hepatocellular carcinoma to anticancer agents". Oncology Reports 36, no. 6 (2016): 3611-3618. https://doi.org/10.3892/or.2016.5188