Activation of estrogen receptors with E2 downregulates peroxisome proliferator-activated receptor γ in hepatocellular carcinoma

  • Authors:
    • Yueh-Min Lin
    • Bharath Kumar Velmurugan
    • Yu-Lan Yeh
    • Chuan  Chou Tu
    • Tsung-Jung Ho
    • Tung  Yuan Lai
    • Chih-Hao  Tsai
    • Fuu  Jen Tsai
    • Chang-Hai Tsai
    • Chih-Yang Huang
  • View Affiliations

  • Published online on: October 10, 2013     https://doi.org/10.3892/or.2013.2793
  • Pages: 3027-3031
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality and occurs more often in men than in women; however, little is known about its underlying molecular mechanisms. The present study investigated the effect of estrogen receptor (ER)α and ERβ on peroxisome proliferator-activated receptor γ (PPARγ) expression in Hep3B cells. We examined PPARγ, ERα and ERβ mRNA and protein expression by RT-PCR and western blotting. In order to determine whether PPARγ plays a central role in HCC, we screened for PPARγ expression in liver cancer patient tissues and differentially differentiated HCC cell lines (HA22T, Huh-7, Hep3B and HepG2). We found that PPARγ expression was highly expressed in liver cancer tissues and in Hep3B cells. Furthermore, overexpression of ERα and ERβ was found to decrease PPARγ expression at the transcriptional as well as at the translational level in a ligand-dependent manner. In summary, the present study demonstrated that both ERα and β were sufficient to inhibit PPARγ and provide a valuable therapeutic option for the treatment of HCC patients.

Introduction

Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide (1) and its frequency is increasing in Southeast Asia, Africa and Western countries. In particular, the mortality rate of HCC in Taiwan has not decreased due to limited treatment options (2,3). HCC occurs more often in men than in women; in addition, males have a poorer prognosis in comparison with females (4). However, little is known about the underlying molecular mechanisms of HCC.

The role of nuclear receptors in HCC development has drawn considerable attention (5). One such example is peroxisome proliferator-activated receptor γ (PPARγ), a ligand-activated transcription factor that is involved in tumor promotion, cellular differentiation and apoptosis (6,7). Several other studies also focused on PPARγ as their target gene to treat various types of cancer, such as colon, thyroid, lung, breast, prostate and liver cancer (8). For example, troglitazone inhibited the growth of human liver cancer cells by inducing apoptosis through caspase-3 activation (9). In breast cancer cells, estrogen receptor (ER)α binds to peroxisome proliferator-activated receptor response element and negatively interferes with PPARγ signaling (10). Similarly, in preadipocytes cells, ERβ overexpression inhibits ligand-mediated PPARγ activity, which further results in a blockade of PPARγ-induced adipocytic gene expression (11).

Expression ratio of ERα and ERβ apparently changes during hepatocarcinogenesis (12). A large body of evidence has shown decreased ERα in HCC patients (13,14); similarly, loss of ERβ expression has been indicated as a common step in the development of colorectal cancer (15). Activation of these ERs controls several biological processes, including cell growth, differentiation and apoptosis. However, the effect of ERα or ERβ on PPARγ expression in HCC is not well studied. In the present study, either ERα or ERβ is overexpressed by transient transfection and then receptor is activated by 17β-estradiol. At the same time, we conducted the assay with 17β-estradiol alone to elucidate whether ligand alone can induce ERα or ERβ expression in ER-negative Hep3B cells. These results showed that ERα or ERβ may act as a tumor suppressor in downregulating PPARγ expression in Hep3B cells and were further accelerated by ligand addition.

Materials and methods

Specimen collection and immunohistochemistry

Written consent was obtained from all patients. Surgical specimens of human liver cancer tissues were obtained by mastectomy from the operating rooms of the Changhua Christian Hospital in Changhua and the China Medical University Hospital in Taichung, Taiwan. Following resection, these specimens were stored at −70°C before being used for the analysis. The tissue biopsy was dried at 58°C overnight, dewaxed in xylene for 40 min and rehydrated in ethanol. Blocking with 3% H202 in 50% methanol/50% phosphate-buffered saline (PBS) and incubated with 5% cosmic calf serum to reduce non-specific staining of the secondary antibody. Tissue sections were incubated overnight at 4°C with PPARγ (1:100). The sections were washed with PBS and incubated for 1 h at room temperature with the peroxidase-conjugated secondary antibody. Immunoreactivity was visualized with 3,3′-diaminobenzidine (DAB) substrate (Roche Diagnostics, Mannheim, Germany). After coloring and rinsing with distilled water, the sections were counterstained slightly with Mayer’s hematoxylin, dehydrated in graded alcohols, cleared in xylene and detected using microscopy (Olympus, Tokyo, Japan).

Cell culture

The Chang liver cell line, HepG2, Hep3B, Huh-7 and HA22T cells were purchased from ATCC. Chang liver cells were grown in DMEM, HepG2 and Hep3B were grown in MEM (Gibco, Grand Island, NY, USA) and Huh-7 and HA22T cells were grown in DMEM. All media were supplemented with 10% fetal bovine serum (FBS) (Biochrom AG, Berlin, Germany) and 1% penicillin streptomycin (Gibco).

Establishment of the double-stable Tet-On/ERα and ERβ Hep3B cell line

The double-stable Tet-On/ERα or ERβ Hep3B cell line, which grows well in the presence of both G418 and hygromycin, was established by plasmid transfection using the Lipofectamine method. Briefly, the primary Tet-On Hep3B cell line was generated by transfecting Hep3B cells with 10 μg Tet-On (Clontech Laboratories, Worcester, MA, USA), a regulator plasmid encoding the G418 resistance gene. The primary Tet-On Hep3B cells were then transfected with 10 μg of pTRE2/ERα or ERβ plasmid encoding the hygromycin resistance gene. Double-stable cells were selected with 700 μg/ml G418 and 100 μg/ml hygromycin and further screened for ERα mRNA using DNA sequencing.

Transfection

Hep3B cells were transfected with a plasmid carrying the ERα and ERβ gene using 10 and 100 μM of Lipofectamine (Invitrogen, Auckland, New Zealand) according to the manufacturer’s guidelines. After 6 h of transfection, MEM supplemented with 10% charcoal/dextran (CD)-FBS (Sigma, St. Louis, MO, USA) was added for 12 h, and MEM containing 1% FBS and antibiotics were added for 6 h. Prior to treatment, the cells were starved in MEM (no phenol red) with 1% antibiotics for 6 h and then replaced with phenol red-free MEM containing 1% FBS and vehicle or 17β-estradiol (E2) (Sigma), doxycycline or fenofibrate (Clontech, Mountain View, CA, USA) for different times. These transfection experiments were repeated three times with consistent results.

Reverse transcription (RT)

Total RNA was extracted using an Ultraspec™ kit (Biotecx, Houston, TX, USA) according to the manufacturer’s instructions. A total of 4 μg of RNA was used for the RT reaction. RT was performed at 37°C for 60 min using 55.5 μl DEPC H2O, 4 μg total RNA, 0.5 μl of RNase inhibitor (40 U/μl) (Promega, Madison, WI, USA), 20 μl of 5X RT buffer, 8 μl of dNTP (2.5 mM), 10 μl of oligo(dT) (5 μM/ml) (Mission Biotech, Taipei, Taiwan) and 2 μl of MMLV reverse transcriptase (200 U/μl) (Promega). The resulting cDNA was added to the PCR mixture containing 9.5 μl of DEPC water, 2.5 μl of 10X PCR buffer (MD Bio, Taipei, Taiwan), 2.5 μl of dNTP (10 mM) (Promega), 2.5 μl of each primer (5 μM), 0.5 μl of Taq (2 U/μl) (MD Bio) and 4 μl of 2.5 mMdNTP mixture.

Western blotting

Cells were lysed at each time-point with lysis buffer [50 mM Tris base (pH 7.4), 0.5 M NaCl, 1 M ethylenediamine-mercaptoethanol (BME), 1% NP-40%, 10% glycerol, Igepal CA-630] (Sigma) and protease inhibitor cocktail tablets (Roche). Proteins were analyzed and separated by 10% SDS-PAGE, transferred to nitrocellulose membranes and probed with antibodies against the following proteins: PPARγ, ERα, ERβ and α-tubulin (Santa Cruz Biotechnology, Santa Cruz, CA, USA). The blots were incubated with peroxidase-conjugated secondary antibody for 1 h. Bands were monitored using western blot chemiluminescence reagent (Santa Cruz Biotechnology).

Statistical analysis

All data are expressed as percentages of the control and mean ± SD. The results are based on three independent experiments. Student’s unpaired t-test was used to compare the differences between groups. Experimental group vs. control group: P-value <0.05 was considered to indicate a statistically significant difference; *P<0.05 and **P<0.01.

Results

PPARγ expression in HCC is significantly increased in tumor tissues compared with surrounding non-tumorous liver, particularly in poorly differentiated tumor compared to well-differentiated tumor (16). To assess the role that PPARγ in liver cancer in vivo, we analyzed tumor and non-tumor patient tissues for PPARγ expression. The majority of normal liver tissues do not express PPARγ, whereas tumor tissues from the liver cancer patients showed a significant increase in PPARγ expression (Fig. 1).

Next, we analyzed the expression of PPARγ in HCC cells in vitro using HepG2, Hep3B, HuH-7 and HA22T cell lines. As shown in Fig. 2A, RT-PCR analysis readily detected the expression of PPARγ mRNA in all cell lines. Western blot analysis did not display exactly same expression pattern when compared with the mRNA expression. Compared with other cell lines used, only Hep3B cells expressed PPARγ protein (Fig. 2B). These results were consistent with the hypothesis that mRNA levels do not necessarily correlate with the protein expression data (17).

Estrogen exerts its biological function by binding to one of two specific ERs, ERα and ERβ. Thus, the level of exogenous ERα and endogenous PPARγ in Hep3B cells transfected with empty vector or ERα expression vectors was examined in the presence of E2. As shown in Fig. 3A, ERα containing Hep3B cells induced ERα mRNA expression and further decreased PPARγ expression. On the other hand, E2 treatment altered the expression level of ERα and PPARγ in ERα overexpressing Hep3B cells. However, in empty vector transfected Hep3B cells, E2 treatment reduced PPARγ mRNA levels without increasing ERα expression. This was further confirmed by western blot analysis (Fig. 3B). We then verified the possibility of ERα in inhibiting PPARγ expression using a stable cell line that expresses ERα. Tet-On/ERα Hep3B cells were treated with a range (0–1.5 μg/ml) of Dox for 24 h and then analyzed for PPARγ expression. As shown in Fig. 3C, a dose-dependent decrease in PPARγ was observed in response to Dox. This was more noticeable at the 0.2 μg/ml concentration.

In order to elucidate whether exogenous expression of ERβ inhibits PPARγ expression, vector or ERβ transfected Hep3B cells were exposed to E2 treatment. No activation of ERβ and PPARγ was observed in cells transfected with empty vector and in E2 exposed cells. Fig. 4 shows that ERβ overexpression plus E2 treatment effectively inhibited PPARγ mRNA and protein expression.

Discussion

The nuclear receptor superfamily (estrogen, thyroid, glucocorticoid receptors and peroxisome proliferator-activated receptors) plays an important role in controlling cellular homeostasis, and administration of its ligand has been effectively used in cancer treatment (1821). The role of PPARγ in tumor development is controversial as fewer studies showed ligand activated PPARγ promotes growth inhibition and apoptosis in human esophageal (22) breast (23) ovarian and liver cancer (24) and other reports showed PPARγ ligand could inhibit growth and metastasis of PPARγ positive cancer cells (25). In the present study, we found a significant increase in PPARγ mRNA expression in HCC tissues compared with non-cancerous tissues. Similarly, in human lung cancer tissues, increased PPARγ expression was observed compared with non-tumor tissues (25). These findings suggest that PPARγ was involved in hepatocarcinogenesis. However, decreased PPARγ expression in tumor tissue has been observed in human esophageal, breast and ovarian cancer (22,26,27). Therefore, a better understanding of the PPARγ mechanism in different cancer tissues is required.

PPARγ expression has been demonstrated in vitro in several cell lines; particularly in liver cancer cell lines, PPARγ mRNA was expressed at various levels (9). Similarly, we analyzed the effects of PPARγ activation in four human HCC cell lines, compared with the HepG2, Huh-7 and HA22T cells, Hep3B cells constitutively express PPARγ expression at the RNA and protein levels. Having observed significant upregulation of PPARγ expression in Hep3B cells, we next conducted experiments to test the potential role of ERs in inhibiting PPARγ expression in Hep3B cells.

Previous studies showed overexpression of ERα inhibits growth of ECV304 and the Ishikawa cell line by decreasing endothelin-1 and VEGF expression (28). Our results, consistent with a previous report (29), showed that ERα binds with PPARγ and functionally interferes with PPARγ signaling in a ligand-dependent manner. Compared with ERα expression, decreased ERβ was found in patients with chronic hepatitis or cirrhosis and in those with HCC. In normal breast cells, ERβ was found to negatively regulate cellular proliferation. Our data are in agreement with these results, showing ERβ overexpression decreased PPARγ expression in an E2-dependent manner. Collectively, the present study provided a basic understanding of ERα and ERβ in PPARγ expression; further studies using these ERs are currently being conducted to elucidate how these ERs control Hep3B cell molecular mechanisms.

Acknowledgements

This research was funded by the China Medical University (grant no. CMU 101-AWARD-04 and CMU 101-S-18).

References

1 

Greten TF, Papendorf F, Bleck JS, et al: Survival rate in patients with hepatocellular carcinoma: a retrospective analysis of 389 patients. Br J Cancer. 92:1862–1868. 2005. View Article : Google Scholar : PubMed/NCBI

2 

El-Serag HB: Hepatocellular carcinoma: recent trends in the United States. Gastroenterology. 127:S27–S34. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Weng CJ, Chau CF, Hsieh YS, Yang SF and Yen GC: Lucidenic acid inhibits PMA-induced invasion of human hepatoma cells through inactivating MAPK/ERK signal transduction pathway and reducing binding activities of NF-κB and AP-1. Carcinogenesis. 29:147–156. 2008.PubMed/NCBI

4 

El-Serag HB, Mason AC and Key C: Trends in survival of patients with hepatocellular carcinoma between 1977 and 1996 in the United States. Hepatology. 33:62–65. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Vacca M, Degirolamo C, Massafra V, et al: Nuclear receptors in regenerating liver and hepatocellular carcinoma. Mol Cell Endocrinol. 368:108–119. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Koeffler HP: Peroxisome proliferator-activated receptor γ and cancers. Clin Cancer Res. 9:1–9. 2003.

7 

Vanden Heuvel JP: Peroxisome proliferator-activated receptors (PPARS) and carcinogenesis. Toxicol Sci. 47:1–8. 1999.PubMed/NCBI

8 

Panigrahy D, Singer S, Shen LQ, et al: PPARγ ligands inhibit primary tumor growth and metastasis by inhibiting angiogenesis. J Clin Invest. 110:923–932. 2002.

9 

Toyoda M, Takagi H, Horiguchi N, et al: A ligand for peroxisome proliferator activated receptor γ inhibits cell growth and induces apoptosis in human liver cancer cells. Gut. 50:563–567. 2002.

10 

Wang X and Kilgore MW: Signal cross-talk between estrogen receptor alpha and beta and the peroxisome proliferator-activated receptor gamma1 in MDA-MB-231 and MCF-7 breast cancer cells. Mol Cell Endocrinol. 194:123–133. 2002. View Article : Google Scholar

11 

Foryst-Ludwig A, Clemenz M, Hohmann S, et al: Metabolic actions of estrogen receptor beta (ERβ) are mediated by a negative cross-talk with PPARγ. PLoS Genet. 4:e10001082008.

12 

Wu X, Subramaniam M, Grygo SB, et al: Estrogen receptor-beta sensitizes breast cancer cells to the anti-estrogenic actions of endoxifen. Breast Cancer Res. 13:R272011. View Article : Google Scholar : PubMed/NCBI

13 

Ng IO, Ng M and Fan ST: Better survival in women with resected hepatocellular carcinoma is not related to tumor proliferation or expression of hormone receptors. Am J Gastroenterol. 92:1355–1358. 1997.PubMed/NCBI

14 

Jonas S, Bechstein WO, Heinze T, et al: Female sex hormone receptor status in advanced hepatocellular carcinoma and outcome after surgical resection. Surgery. 121:456–461. 1997. View Article : Google Scholar : PubMed/NCBI

15 

Kennelly R, Kavanagh DO, Hogan AM and Winter DC: Oestrogen and the colon: potential mechanisms for cancer prevention. Lancet Oncol. 9:385–391. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Yu J, Shen B, Chu ES, et al: Inhibitory role of peroxisome proliferator-activated receptor gamma in hepatocarcinogenesis in mice and in vitro. Hepatology. 51:2008–2019. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Borbath I and Horsmans Y: The role of PPARγ in hepatocellular carcinoma. PPAR Res. 2008:2095202008.

18 

Li MY, Deng H, Zhao JM, Dai D and Tan XY: Peroxisome proliferator-activated receptor gamma ligands inhibit cell growth and induce apoptosis in human liver cancer BEL-7402 cells. World J Gastroenterol. 9:1683–1688. 2003.PubMed/NCBI

19 

Lovat PE, Oliverio S, Ranalli M, et al: GADD153 and 12-lipoxygenase mediate fenretinide-induced apoptosis of neuroblastoma. Cancer Res. 62:5158–5167. 2002.PubMed/NCBI

20 

Galbiati E, Caruso PL, Amari G, et al: Pharmacological actions of a novel, potent, tissue-selective benzopyran estrogen. J Pharmacol Exp Ther. 303:196–203. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Chen GG, Lee JF, Wang SH, Chan UP, Ip PC and Lau WY: Apoptosis induced by activation of peroxisome-proliferator activated receptor-gamma is associated with Bcl-2 and NF-kappaB in human colon cancer. Life Sci. 70:2631–2646. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Terashita Y, Sasaki H, Haruki N, et al: Decreased peroxisome proliferator-activated receptor gamma gene expression is correlated with poor prognosis in patients with esophageal cancer. Jpn J Clin Oncol. 32:238–243. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Cui Y, Miyoshi K, Claudio E, et al: Loss of the peroxisome proliferation-activated receptor gamma (PPARγ) does not affect mammary development and propensity for tumor formation but leads to reduced fertility. J Biol Chem. 277:17830–17835. 2002.

24 

Yu J, Qiao L, Zimmermann L, et al: Troglitazone inhibits tumor growth in hepatocellular carcinoma in vitro and in vivo. Hepatology. 43:134–143. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Li MY, Hsin MK, Yip J, Mok TS, Underwood MJ and Chen GG: PPARgamma activation extinguishes smoking carcinogen by inhibiting NNK-mediated proliferation. Am J Respir Cell Mol Biol. 42:113–122. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Badawi AF and Badr MZ: Expression of cyclooxygenase-2 and peroxisome proliferator-activated receptor-γ and levels of prostaglandin E2 and 15-deoxy-Δ12,14-prostaglandin J2 in human breast cancer and metastasis. Int J Cancer. 103:84–90. 2003.

27 

Sakamoto A, Yokoyama Y, Umemoto M, et al: Clinical implication of expression of cyclooxygenase-2 and peroxisome proliferator activated-receptor gamma in epithelial ovarian tumours. Br J Cancer. 91:633–638. 2004.PubMed/NCBI

28 

Upla P, Marjomaki V, Kankaanpaa P, et al: Clustering induces a lateral redistribution of α2β1 integrin from membrane rafts to caveolae and subsequent protein kinase C-dependent internalization. Mol Biol Cell. 15:625–636. 2004.

29 

Bonofiglio D, Gabriele S, Aquila S, et al: Estrogen receptor α binds to peroxisome proliferator-activated receptor response element and negatively interferes with peroxisome proliferator-activated receptor γ signaling in breast cancer cells. Clin Cancer Res. 11:6139–6147. 2005.

Related Articles

Journal Cover

December 2013
Volume 30 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
Lin Y, Velmurugan BK, Yeh Y, Tu CC, Ho T, Lai TY, Tsai C, Tsai FJ, Tsai C, Huang C, Huang C, et al: Activation of estrogen receptors with E2 downregulates peroxisome proliferator-activated receptor γ in hepatocellular carcinoma. Oncol Rep 30: 3027-3031, 2013
APA
Lin, Y., Velmurugan, B.K., Yeh, Y., Tu, C.C., Ho, T., Lai, T.Y. ... Huang, C. (2013). Activation of estrogen receptors with E2 downregulates peroxisome proliferator-activated receptor γ in hepatocellular carcinoma. Oncology Reports, 30, 3027-3031. https://doi.org/10.3892/or.2013.2793
MLA
Lin, Y., Velmurugan, B. K., Yeh, Y., Tu, C. C., Ho, T., Lai, T. Y., Tsai, C., Tsai, F. J., Tsai, C., Huang, C."Activation of estrogen receptors with E2 downregulates peroxisome proliferator-activated receptor γ in hepatocellular carcinoma". Oncology Reports 30.6 (2013): 3027-3031.
Chicago
Lin, Y., Velmurugan, B. K., Yeh, Y., Tu, C. C., Ho, T., Lai, T. Y., Tsai, C., Tsai, F. J., Tsai, C., Huang, C."Activation of estrogen receptors with E2 downregulates peroxisome proliferator-activated receptor γ in hepatocellular carcinoma". Oncology Reports 30, no. 6 (2013): 3027-3031. https://doi.org/10.3892/or.2013.2793