Saikosaponin‑d suppresses the expression of cyclooxygenase‑2 through the phospho‑signal transducer and activator of transcription 3/hypoxia‑inducible factor‑1α pathway in hepatocellular carcinoma cells

  • Authors:
    • Shuixiang He
    • Guifang Lu
    • Helei Hou
    • Zhenjun Zhao
    • Zhanfang Zhu
    • Xinlan Lu
    • Jinghong Chen
    • Zhilun Wang
  • View Affiliations

  • Published online on: September 16, 2014     https://doi.org/10.3892/mmr.2014.2574
  • Pages: 2556-2562
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is one of the most common malignancies and accounts for ~6% of all types of human cancer worldwide, particularly in Asia. The incidence and mortality rates in the USA have also rapidly increased. Saikosaponin‑d (SSD), a saponin derivative extracted from several species of Bupleurum (Umbelliferae), possesses unique biological activities, including anti‑inflammatory, antihepatitic and immunomodulatory effects. Our previous studies have demonstrated that SSD inhibits the proliferation and induces the apoptosis of HCC SMMC‑7721 cells by downregulating the expression of cyclooxygenase (COX)‑2 and decreasing the production of prostaglandin E2. However, the specific mechanism underlying how SSD controls the expression of COX‑2 remains to be elucidated. In the present study, it was demonstrated that hypoxia inducible factor‑1α (HIF‑1α) was responsible for the expression of COX‑2 under hypoxic conditions in HCC cells, and the activation of signal transducer and activator of transcription 3 (STAT3) was required for the expression of HIF‑1α. SSD treatment inhibited STAT3 activation [phosphorylation of STAT3 (p‑STAT3)], reduced the protein level of HIF‑1α and decreased the expression of COX‑2. These results suggested that SSD may target HCC cells by suppressing the expression of COX‑2 through the p‑STAT3/HIF‑1α pathway.

Introduction

Hepatocellular carcinoma (HCC), accounting for 70–85% of the total liver cancer burden, is one of the most common malignancies and is the third leading cause of cancer mortality worldwide, with an estimated >500,000 new cases per year (1,2). At present, curative therapies, including resection, liver transplantation and ablation, provide effective treatment for only a small number of patients presenting with early stage HCC in the clinic. The majority of patients with intermediate-advanced HCC are only eligible for the mainstream palliative treatments, including transarterial chemoembolization and systemic therapy with molecular targeted drugs (3). However, therapies against liver cancer to date have not been completely effective. In this context, the development of new, effective therapeutic approaches for liver cancer remains one of the most challenging goals in cancer research.

Numerous traditional Chinese plants have been identified to possess biological activities with potential therapeutic applications. Saikosaponin-d (SSD), a saponin derivative extracted from several species of Bupleurum (Umbelliferae), has been traditionally used in the treatment of infectious diseases due to its anti-inflammatory, antipyretic and analgesic effects (4,5). Previous studies have demonstrated that SSD also has hepatoprotective, antifibrotic (6,7) and immunomodulatory (8,9) activities. Furthermore, traditional use and scientific studies have suggested that SSD is a potential candidate as an anticancer agent (10,11), which has been demonstrated to have anti-proliferative and apoptotic effects on various cancer cells, including human leukemia cancer, non-small cell lung cancer (12) and hepatic cancer (13). Our previous study demonstrated that SSD inhibits the proliferation and induces the apoptosis of HCC SMMC-7721 cells by downregulating cyclooxygenase (COX)-2 at the mRNA and protein level and inhibiting the production of prostaglandin E2 (PGE2) (14). However, the specific mechanism underlying how SSD controls COX-2 expression remains to be elucidated.

COX-2, a key inducible enzyme in prostanoid biosynthesis, is overexpressed in solid malignancies, including colon, prostate, breast and HCC (15). A significant negative correlation between the overexpression of COX-2 and the survival rates of patients in various types of cancer has been reported in retrospective studies (1618). Inhibiting the activity or expression of COX-2 has shown promise for tumor therapy in animal models and cancer patients (19,20). In HCC patients, the protein expression of COX-2 correlates well with the differentiation grades, suggesting that abnormal COX-2 expression has an important effect in hepatocarcinogenesis (21). It is well established that non-steroidal anti-inflammatory drugs (NSAIDs) have anti-tumor effects by acting on COX-2 (22). SSD has a similar pharmacological activity to NSAIDs, and it has been documented that SSD inhibits HCC cell proliferation by modulating COX-2 expression (14). However, how SSD regulates the expression of COX-2 remains to be elucidated.

Materials and methods

Cell culture and reagents

The liver cancer cell lines SMMC-7721 and HepG2, obtained from the Transform Medical Center of Xi’an Jiaotong University (Xi’an, China), were cultured in RPMI-1640 medium (Invitrogen Life Technologies, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (HyClone, Logan, UT, USA) and 1% penicillin/streptomycin, and incubated at 37°C in a humidified 5% CO2 atmosphere. The Janus kinase 2 (JAK2) selective inhibitor AG-490, hypoxia simulator cobalt chloride (CoCl2), mammalian target of rapamycin (mTOR) and SSD were obtained from Sigma (Poole, UK). AG-490, rapamycin and SSD were dissolved in dimethyl sulfoxide (DMSO; Sigma-Aldrich, St. Louis, MO, USA), and interleukin-6 (IL-6) was dissolved in acetic acid (Sigma-Aldrich). For all experiments, final concentrations of the tested regents were prepared on the day of assessment by diluting the stock with RPMI-1640 medium and the final concentration of DMSO was <0.1%, which was not considered to be harmful to the cells.

Cell proliferation assay

The effect of SSD on cell proliferation was examined using an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. SMMC-7721 cells were plated in 96-well plates at a density of 5×103 cells per well and were allowed to grow to 70% confluence. After 24 h, the cells were randomly separated into four groups and were treated with SSD at 2.5, 5.0, 10 and 15 μg/ml, respectively. After 0, 24, 48 and 72 h, 20 μl of MTT test solution, which was freshly prepared, was added to each well. After 4 h incubation, the supernatant was discarded and 150 μl DMSO was added to dissolve the crystal. All analyses were performed in triplicate. The absorbance was measured on an ELISA reader (Thermo Fisher Scientific, Waltham, MA, USA) at a test wavelength of 490 nm. Proliferation inhibition rate (%) = (control well A490 - experiment well A490) / control well A490 × 100%.

Western blot analysis

Tumor cells were plated in 100 mm cell culture dishes (Nunc A/S, Roskilde, Denmark) with ~300×104 cells per dish. When cells grew to 60–70% confluence, they were randomly separated into different groups to be treated with either CoCl2, CoCl2 + rapamycin, CoCl2 + AG490 or CoCl2 + SSD. After 24 h, whole cell protein extracts were prepared by lysing cells with radioimmunoprecipitation assay lysis buffer supplemented with a protease inhibitor cocktail (Roche Diagnostics, Mannheim, Germany) and phosphate inhibitor PhosStop (Roche Diagnostics). Protein concentration was quantified using the Bradford method. For western blotting, total cell lysates (~100 μg per lane) were subjected to SDS-PAGE. The protein was then transferred onto polyvinylidene difluoride membranes (Millipore Corp., Billerica, MA, USA) using semi-dry transfer instruments (Bio-Rad Laboratories, Hercules, CA, USA) at 15 V for 30 min. The membranes were incubated with blocking buffer (0.05% Tween 20 with 5% nonfat milk) for 1 h at room temperature followed by anti-COX-2, anti-HIF-1α or anti-phospho-STAT3 primary rabbit anti-human monoclonal antibody (Cell Signaling Technology, Inc., Danvers, MA, USA; 1:1,000) dilution buffer overnight at 4°C. Following washing three times with washing buffer (blocking buffer without 5% nonfat milk) for 10 min each time, the membranes were incubated with horseradish peroxidase-labeled goat anti-rabbit secondary antibody (polyclonal; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA; 1:5,000) for 1 h at 37°C. The membranes were washed again and detection was performed using an enhanced chemiluminescence western blotting detection system (Pierce Biotechnology, Inc., Rockford, IL, USA).

Reverse transcription-polymerase chain reaction (RT-PCR)

Total cellular RNA was isolated from each of the experimental groups using TRIzol solution (Invitrogen Life Technologies). RT was performed on RNA samples, followed by PCR amplification. For RT, 1.0 μg of the RNA sample was added to 20 μl of RT reaction mixture (Fermentas, Waltham, MA, USA). The reaction was performed by treating the samples at 65°C for 5 min, at 42°C for 60 min and at 70°C for 5 min. PCR was conducted using the following primers specific for each of the target genes: HIF-1α, sense 5′-CATTAGAAAGCAGTTCCGCAAGC-3′ and antisense 5′-CAGTGGTAGTGGTGGCATTAGC-3′; COX-2, sense 5′-AGTATCACAGGCTTCCATTGACCAG-3′ and antisense 5′-CCACAGCATCGATGTCACCATAG-3′; β-actin, sense 5′-ATCGTGCGTGACATTAAGGAGAAG-3′ and antisense 5′-AGGAAGGAAGGCTGGAAGAGTG-3′. The PCR was initiated in a thermal cycle programmed at 94°C for 5 min, 94°C for 30 sec, 58°C for 30 sec, 72°C for 60 sec, and amplified for 30 cycles with HIF-1α and β-actin, and 35 cycles with COX-2. The amplified products were visualized on 1.5% agarose gels.

Immunocytochemical staining

Immunocytochemical staining was performed on the coverslips obtained from the experimental groups. The antibodies against HIF-1α and COX-2 were purchased from Beijing Biosynthesis Biotechnology Co., Ltd (Beijing, China) and used according to the manufacturer’s instructions. Briefly, the coverslips were incubated for 20 min in 3% H2O2. Following washing with phosphate-buffer saline, the coverslips were incubated with the appropriately diluted first antibody (1:400) at 4°C overnight in a humid chamber, followed by treatment with biotinylated immunoglobulin for 12 min after washing, and then with streptavidin/horseradish peroxidase for 12 min at 37°C. The color reaction was developed using diaminobenzidine working solution (Tiangen Biotech Co., Ltd., Beijing, China) for 3–5 min and counterstained with hematoxylin.

Statistical analysis

The results were analyzed for statistical significance using Student’s t-test between the incubation conditions of normoxia and hypoxia under multiple exposure conditions using SPSS 16.0 software (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

SSD inhibits SMMC-7721 cell proliferation and alters its cell morphology

MTT assay was used to detect the effect of SSD on SMMC-7721 cell proliferation. SMMC-7721 cells were treated with SSD at various concentrations (2.5, 5, 10 and 15 μg/ml) for 0, 24, 48 and 72 h. The results demonstrated that the growth inhibitory effect of SSD on SMMC-7721 cells was in a time- and dose-dependent manner (Fig. 1). Following treatment with 10 μg/ml SSD, SMMC-7721 cell proliferation activity was significantly reduced. Morphologically, the cells detached from the bottle and became round. In addition, a transparent vacuolar structure and pyknosis of the nucleus was observed (Fig. 2). This phenomenon was most clear at 72 h.

HIF-1α is necessary for COX-2 expression in HCC cells

Hypoxia commonly occurs in solid tumors and HIF-1α is critical in the hypoxia adaptation process. Several studies have investigated the importance of COX-2 in tumorigenesis and analysis has identified COX-2 as a direct target for HIF-1α in colorectal tumor cells (12,13). Furthermore, COX-2 upregulation represents a pivotal cellular adaptive response to hypoxia with implications for colorectal tumor cell survival and angiogenesis. Therefore, the present study aimed to determine whether COX-2 expression was controlled by HIF-1α in HCC. CoCl2 was able to inhibit the degradation of HIF-1α under normoxic conditions and is used to simulate hypoxia in experiments (23). SMMC-7721 cells were subjected to CoCl2-stimulated hypoxia and protein extracts were prepared over time at several concentrations of CoCl2. Western blotting revealed that HIF-1α and COX-2 protein levels were rapidly induced by CoCl2 in a time- and dose-dependent manner (Fig. 3A). The gene expression of HIF-1α and COX-2 was also detected. Although HIF-1α mRNA levels did not alter with different concentrations of CoCl2, COX-2 mRNA levels increased under CoCl2-stimulated hypoxic conditions in a dose-dependent manner in RT-PCR analysis (Fig. 3B).

To further examine the effect of HIF-1α on COX-2 induction, SMMC-7721 cells were treated with rapamycin, a reagent which could inhibit the synthesis of HIF-1α. Western blotting and RT-PCR analysis demonstrated that rapamycin eliminated COX-2 upregulation at the protein and mRNA levels under CoCl2-stimulated hypoxia conditions (Fig. 3C and D). This suggested that HIF-1α was an upstream regulator of COX-2 expression in HCC.

Inhibition of STAT3 phosphorylation reduces the expression of HIF-1α and COX-2

The activated form of STAT3 (p-STAT3) is highly expressed in several malignancies, which has been demonstrated to induce HIF-1α protein synthesis in human breast tumor MCF-7 cells (24). However, the association between COX-2, HIF-1α and p-STAT3 in HCC cells remains to be elucidated. In order to determine the association between p-STAT3 and HIF-1α/COX-2, SMMC-7721 cells were treated with AG-490 for 30 min prior to the addition of CoCl2. AG-490 is the selective inhibitor of JAK2, which can inhibit the activation of STAT3 (25). The results demonstrated that AG-490 resulted in the downregulation of HIF-1α and COX-2 at the protein level under the hypoxic conditions simulated by CoCl2 (Fig. 4A). In order to confirm this effect, this was repeated on HepG2 cells and the result was the same as that observed in SMMC-7721 cells (Fig. 4B).

Effects of SSD on the protein expression of p-STAT3, HIF-1α and COX-2

In order to determine the mechanism of SSD targeting in HCC cells, the expression of COX-2, HIF-1α and p-STAT3 was determined by immunocytochemistry following SSD treatment. The results suggested that p-STAT3 staining in SMMC-7721 tumor cells demonstrated nuclear localization, with HIF-1α located in the cytoplasm and particularly in the nucleus, and COX-2 expressed in the cytoplasm and nuclear membrane. SSD not only significantly reduced the expression of HIF-1α and COX-2 induced by CoCl2, but also deceased the expression of p-STAT3 and COX-2 induced by IL-6 (Fig. 5). These results were partially verified by western blotting, which indicated that SSD inhibited the protein expression of COX-2, HIF-1α and p-STAT3 (Fig. 6).

Discussion

HCC is one of the most common types of malignancy worldwide, the incidence and mortality rate of HCC are extremely high in Asia and have also increased rapidly in the United States (1,2). Due to the occult onset of HCC, the majority of HCC patients are at an advanced stage when diagnosed and to date there remains no completely effective therapy for HCC (3). Numerous active compounds extracted from traditional plants, including SSD, have been demonstrated to have anti-tumor activities (10,11). SSD could not only inhibit growth and differentiation of human leukemia cells (26) and glioma cells (27), but was also be able to increase the radiosensitivity of HCC SMMC-7721 cells by adjusting the G0/G1 and G2/M checkpoints of the cell cycle (28). The present study demonstrated that SSD inhibited COX-2 expression through the STAT3/HIF-1α signaling pathway, which may be the specific antitumor mechanism of SSD.

COX, the key enzyme for prostanoid biosynthesis, has two isoforms: COX-1 and COX-2. COX-1 is constitutively expressed in several tissues and cell types, whereas COX-2 is an inducible enzyme expressed only in response to certain stimuli. COX-2 is overexpressed in a subset of malignant tumors and accumulating evidence suggests that COX-2 may be important in tumorigenesis through multiple mechanisms (15). Previous studies have confirmed that COX-2 is not only overexpressed in HCC, but also correlates well with the differentiation grades of HCC (21). Our previous study (14) found that SSD inhibited COX-2 expression in HCC SMMC-7721 cells, which confirmed the hypothesis that SSD, with similar pharmacological activities as NASIDs, could inhibit SMMC-7721 proliferation through the COX-2 pathway.

The association between tumors and microenvironments has attracted more attention (29). The hypoxic microenvironment, one of the basic features in solid tumors, characterized by deficiency in oxygen and nutrients, leads to epigenetic and genetic adaptation of clones and an increase in invasiveness and metastasis (30). These hypoxic adaptations, including increasing vascularization, activation of proto-oncogenes, increasing glucose transportation and inducing glycolytic enzymes and various apoptotic-related genes make the tumors more difficult to treat and confers increased resistance to chemotherapy and radiotherapy (31). It is believed that the HIF-1 complex, composed of a heterodimer pair of HIF-1α and HIF-1β is important in mediating these adaptations. At present, HIF-1α has emerged as an important transcription factor in cancer biology and is expressed in the early stages of several types of human malignant tumor, including HCC (32). Csiki et al (33) demonstrated that COX-2 is upregulated in hypoxic lung cancer cells in an HIF-1-dependent manner. Another study provided the first evidence, to the best of our knowledge, demonstrating that HIF-1 directly binds a specific hormone response element located at the COX-2 promoter (34). Dai et al (35) demonstrated a positive correlation between HIF-1α and COX-2 in HCC. The present study found that hypoxia, imitated by CoCl2, could induce the expression of HIF-1α accompanied by the protein level of COX-2 in SMMC-7721 cells. Rapamycin, a selective inhibitor of mTOR, could inhibit the expression of HIF-1α and the COX-2 protein, and SSD had a similar effect. The results of the present study were consistent with previous studies, demonstrating that HIF-1α was obligatory for COX-2 expression in HCC cells and is possibly an important upstream factor for COX-2.

The level of HIF-1α can be regulated not only by hypoxia through a ubiquitin-proteasome pathway, but can also be modulated by several other pathways. The JAK/STAT3 pathway appears to be important in modulating HIF-1α expression. Activated STAT3 can increase HIF-1α protein levels by inhibiting HIF-1α degradation and accelerating its de novo synthesis in ischemic rat kidneys and hypoxic human renal carcinoma cells (36). STAT3 knockout eliminates estrogen receptor-α-induced HIF-1α and subsequent vascular endothelial growth factor (VEGF) production (37). In human breast cancer MCF-7 cells, STAT3 regulates HIF-1α, and targeting STAT3 with siRNA knockdown inhibits CoCl2-mediated HIF-1α nuclear accumulation and recruitment on the VEGF promoter (38). The present study demonstrated that activated STAT3 was involved in the expression of HIF-1α in HCC cells, and p-STAT3 was able to increase HIF-1α protein levels but not mRNA levels. This suggested that activation of STAT3 modulated HIF-1α expression through transcriptional or posttranscriptional mechanisms in HCC. The results of the present study suggested that p-STAT3 may be the upstream regulator of HIF-1α and COX-2 and that there was a p-STAT3/HIF-1α/COX-2 signal transduction pathway in HCC SMMC-7721 cells. In the present study, SSD inhibited SMMC-7721 growth accompanied by a reduction in the expression of p-STAT3, HIF-1α and COX-2. This indicated that SSD may suppress HCC SMMC-7721 proliferation by inhibiting the expression of COX-2 through the p-STAT3/HIF-1α signaling pathway.

In conclusion, the present study provided primary evidence that HIF-1α promoted COX-2 expression under hypoxic conditions in HCC cells and HIF-1α was induced by activated STAT3. SSD may suppress HCC SMMC-7721 cell proliferation by inhibiting the expression of COX-2 through the p-STAT3/HIF-1α signaling pathway.

Acknowledgements

The authors would like to thank the staff of the Endemic Laboratory, Xi’an Jiaotong University for their technical assistance in these studies. This study was supported by the Chinese National Natural Science Foundation (grant no. 30771895).

References

1 

Montalto G, Cervello M, Giannitrapani L, Dantona F, Terranova A and Castagnetta LA: Epidemiology, risk factors, and natural history of hepatocellular carcinoma. Ann NY Acad Sci. 963:3–20. 2002.PubMed/NCBI

2 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar

3 

El-Serag HB, Marrero JA, Rudolph L and Reddy KR: Diagnosis and treatment of hepatocellular carcinoma. Gastroenterology. 134:1752–1763. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Lu CN, Yuan ZG, Zhang XL, et al: Saikosaponin a and its epimer saikosaponin d exhibit anti-inflammatory activity by suppressing activation of NF-κB signaling pathway. Int Immunopharmacol. 14:121–126. 2012.PubMed/NCBI

5 

Hattori T, Nishimura H, Kase Y and Takeda S: Saireito and saikosaponin D prevent urinary protein excretion via glucocorticoid receptor in adrenalectomized WKY rats with heterologous-phase anti-GBM nephritis. Nephron Physiol. 109:19–27. 2008. View Article : Google Scholar

6 

Fan J, Li X, Li P, et al: Saikosaponin-d attenuates the development of liver fibrosis by preventing hepatocyte injury. Biochem Cell Biol. 85:189–195. 2007.PubMed/NCBI

7 

Dang S, Wang B, Cheng Y, Song P, Liu Z and Li Z: Inhibitory effects of saikosaponin-d on CCl4-induced hepatic fibrogenesis in rats. World J Gastroenterol. 13:557–563. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Kato M, Pu M, Isobe K, et al: Characterization of the immunoregulatory action of saikosaponin-d. Cell Immunol. 159:15–25. 1994. View Article : Google Scholar

9 

Wong VK, Zhou H, Cheung SS, Li T and Liu L: Mechanistic study of saikosaponin-d (Ssd) on suppression of murine T lymphocyte activation. J Cell Biochem. 107:303–315. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Man S, Gao W, Zhang Y, Huang L and Liu C: Chemical study and medical application of saponins as anti-cancer agents. Fitoterapia. 81:703–714. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Bachran C, Bachran S, Sutherland M, Bachran D and Fuchs H: Saponins in tumor therapy. Mini Rev Med Chem. 8:575–584. 2008. View Article : Google Scholar

12 

Hsu Y, Kuo P and Lin C: The proliferative inhibition and apoptotic mechanism of Saikosaponin D in human non-small cell lung cancer A549 cells. Life Sci. 75:1231–1242. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Hsu Y, Kuo P, Chiang L and Lin C: Involvement of p53, nuclear factor kappaB and Fas/Fas ligand in induction of apoptosis and cell cycle arrest by saikosaponin d in human hepatoma cell lines. Cancer Lett. 213:213–221. 2004. View Article : Google Scholar : PubMed/NCBI

14 

He SX, Luo JY, Zhao G, et al: Effect of saikosaponins-d on cyclooxygenase-2 expression of human hepatocellular carcinoma cell line smmc-7721. Zhonghua Gan Zang Bing Za Zhi. 14:712–714. 2006.(In Chinese).

15 

Ristimaki A: Cyclooxygenase 2: from inflammation to carcinogenesis. Novartis Found Symp. 256:215–269. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Bin W, He W, Feng Z, et al: Prognostic relevance of cyclooxygenase-2 (COX-2) expression in Chinese patients with prostate cancer. Acta Histochem. 113:131–136. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Matsubayashi H, Infante JR, Winter JM, et al: Tumor COX-2 expression and prognosis of patients with resectable pancreatic cancer. Cancer Biol Ther. 6:1569–1575. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Mascaux C, Martin B, Paesmans M, et al: Has Cox-2 a prognostic role in non-small-cell lung cancer? A systematic review of the literature with meta-analysis of the survival results. Br J Cancer. 95:139–145. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Harris RE: Cyclooxygenase-2 (COX-2) blockade in the chemoprevention of cancers of the colon, breast, prostate, and lung. Inflammopharmacology. 17:55–67. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Harris RE, Beebe-Donk J and Alshafie GA: Cancer chemoprevention by cyclooxygenase 2 (COX-2) blockade: results of case control studies. Subcell Biochem. 42:193–212. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Bae SH, Jung ES, Park YM, et al: Expression of cyclooxygenase-2 (COX-2) in hepatocellular carcinoma and growth inhibition of hepatoma cell lines by a COX-2 inhibitor, NS-398. Clin Cancer Res. 7:1410–1418. 2001.PubMed/NCBI

22 

Cha YI and DuBois RN: NSAIDs and cancer prevention: Targets downstream of COX-2. Annu Rev Med. 58:239–252. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Yuan Y, Hilliard G, Ferguson T and Millhorn DE: Cobalt inhibits the interaction between hypoxia-inducible factor-alpha and von Hippel-Lindau protein by direct binding to hypoxia-inducible factor-alpha. J Biol Chem. 278:15911–15916. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Xu Q, Briggs J, Park S, et al: Targeting Stat3 blocks both HIF-1 and VEGF expression induced by multiple oncogenic growth signaling pathways. Oncogene. 24(36): 5552–5560. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Yu JH, Kim KH and Kim H: Suppression of IL-1beta expression by the Jak 2 inhibitor AG490 in cerulein-stimulated pancreatic acinar cells. Biochem Pharmacol. 72:1555–1562. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Bu S, Xu J and Sun J: Effect of saikosaponin-d on up-regulating GR mRNA expression and inhibiting cell growth in human leukemia cells. Zhongguo Zhong Xi Yi Jie He Za Zhi. 20:350–352. 2000.(In Chinese).

27 

Tsai YJ, Chen I, Horng LY and Wu RT: Induction of differentiation in rat C6 glioma cells with Saikosaponins. Phytother Res. 16:117–121. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Wang B, Dai Z, Wang X, et al: Saikosaponin-d increases the radiosensitivity of smmc-7721 hepatocellular carcinoma cells by adjusting the g0/g1 and g2/m checkpoints of the cell cycle. BMC Complement Altern Med. 13:2632013. View Article : Google Scholar : PubMed/NCBI

29 

Whiteside TL: The tumor microenvironment and its role in promoting tumor growth. Oncogene. 27:5904–5912. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Bartkowiak K, Riethdorf S and Pantel K: The interrelating dynamics of hypoxic tumor microenvironments and cancer cell phenotypes in cancer metastasis. Cancer Microenviron. 5:59–72. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Ke Q and Costa M: Hypoxia-inducible factor-1 (HIF-1). Mol Pharmacol. 70:1469–1480. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Zheng S, Chen X, Yin X and Zhang B: Prognostic significance of HIF-1α expression in hepatocellular carcinoma: a meta-analysis. PLoS One. 8:e657532013.

33 

Csiki I, Yanagisawa K, Haruki N, et al: Thioredoxin-1 modulates transcription of cyclooxygenase-2 via hypoxia-inducible factor-1alpha in non-small cell lung cancer. Cancer Res. 66:143–150. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Kaidi A, Qualtrough D, Williams AC and Paraskeva C: Direct transcriptional up-regulation of cyclooxygenase-2 by hypoxia-inducible factor (HIF)-1 promotes colorectal tumor cell survival and enhances HIF-1 transcriptional activity during hypoxia. Cancer Res. 66:6683–6691. 2006. View Article : Google Scholar

35 

Dai C, Gao Q, Qiu S, et al: Hypoxia-inducible factor-1 alpha, in association with inflammation, angiogenesis and MYC, is a critical prognostic factor in patients with HCC after surgery. BMC Cancer. 9:4182009. View Article : Google Scholar : PubMed/NCBI

36 

Jung JE, Lee HG, Cho IH, et al: STAT3 is a potential modulator of HIF-1-mediated VEGF expression in human renal carcinoma cells. FASEB J. 19:1296–1298. 2005.PubMed/NCBI

37 

Wang M, Tan J, Coffey A, Fehrenbacher J, Weil BR and Meldrum DR: Signal transducer and activator of transcription 3-stimulated hypoxia inducible factor-1 alpha mediates estrogen receptor-alpha-induced mesenchymal stem cell vascular endothelial growth factor production. J Thorac Cardiovasc Surg. 138:163–171. 2009. View Article : Google Scholar

38 

Cascio S, D’Andrea A, Ferla R, et al: miR-20b modulates VEGF expression by targeting HIF-1 alpha and STAT3 in MCF-7 breast cancer cells. J Cell Physiol. 224:242–249. 2010.PubMed/NCBI

Related Articles

Journal Cover

November-2014
Volume 10 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He S, Lu G, Hou H, Zhao Z, Zhu Z, Lu X, Chen J and Wang Z: Saikosaponin‑d suppresses the expression of cyclooxygenase‑2 through the phospho‑signal transducer and activator of transcription 3/hypoxia‑inducible factor‑1α pathway in hepatocellular carcinoma cells. Mol Med Rep 10: 2556-2562, 2014
APA
He, S., Lu, G., Hou, H., Zhao, Z., Zhu, Z., Lu, X. ... Wang, Z. (2014). Saikosaponin‑d suppresses the expression of cyclooxygenase‑2 through the phospho‑signal transducer and activator of transcription 3/hypoxia‑inducible factor‑1α pathway in hepatocellular carcinoma cells. Molecular Medicine Reports, 10, 2556-2562. https://doi.org/10.3892/mmr.2014.2574
MLA
He, S., Lu, G., Hou, H., Zhao, Z., Zhu, Z., Lu, X., Chen, J., Wang, Z."Saikosaponin‑d suppresses the expression of cyclooxygenase‑2 through the phospho‑signal transducer and activator of transcription 3/hypoxia‑inducible factor‑1α pathway in hepatocellular carcinoma cells". Molecular Medicine Reports 10.5 (2014): 2556-2562.
Chicago
He, S., Lu, G., Hou, H., Zhao, Z., Zhu, Z., Lu, X., Chen, J., Wang, Z."Saikosaponin‑d suppresses the expression of cyclooxygenase‑2 through the phospho‑signal transducer and activator of transcription 3/hypoxia‑inducible factor‑1α pathway in hepatocellular carcinoma cells". Molecular Medicine Reports 10, no. 5 (2014): 2556-2562. https://doi.org/10.3892/mmr.2014.2574