Skip to main content

Cellular effects induced by 17-β-estradiol to reduce the survival of renal cell carcinoma cells

Abstract

Background

Renal cell carcinoma (RCC) is an adult malignancy with 2:1 men-to-women ratio, which implies the possible role of sex hormones in RCC carcinogenesis. One of the predominant sex hormones in women before menopause, 17-β-estradiol (or E2), may regulate RCC growth by cellular mechanisms that are still not fully understood.

Methods

The expression levels of E2 receptors (ER1 and ER2) were determined in different RCC cell lines. The DNA damage response induced by E2 was determined by a DNA double-strand break marker γH2AX. To study the possible effect of E2 on oxidative stress response, RCC cells were stained with 2,7-dichlorofluorescein diacetate and analyzed by flow cytometry. Upregulation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) ser40 phosphorylation in response to oxidative stress was detected by immunoblotting. Finally, annexin V/propidium iodide (PI) double staining assay was used to determine E2-induced cellular apoptosis.

Results

Variable expression of ER1 and ER2 were found in the RCC cell lines studied (786-O, A498, and ACHN), in which ACHN and A498 showed highest and lowest ER expression, respectively. In A498 cells, E2 induced DNA double-strand breaks with positive staining of γH2AX. On the other hand, the level of reactive oxidative species were elevated in ACHN cells after E2 treatment. The E2-induced oxidative stress also induced the Ser40 phosphorylation and nuclear translocation of Nrf2. Finally, we also demonstrated that E2 induced apoptosis as revealed by annexin V/PI double staining.

Conclusions

In this study, we demonstrated the cellular effects of E2 on DNA repair, ROS production as well as Nrf2 activation, and apoptosis in RCC cell lines. Together these cellular alterations may contribute to the reduced viability of RCC cells following E2 treatment.

Background

Renal cell carcinoma (RCC) is the most common solid neoplasm of the kidney in adult human. This kidney cancer exhibits a 2:1 male-to-female ratio with a better survival rate in younger women [16]. Thus, there appear to be hormonal effects on the development, progression, and treatment of RCC [31, 33].

In RCC, altered chemosensitivity mediated by 17-β-estradiol (E2) has been reported, so adjuvant therapy with E2 is possible as in other human diseases [24, 45]. The molecular mechanisms of the action of E2 in RCC cells have been reported in recent years [8, 10, 13, 43, 45]. However, diverse genetic alterations are linked to renal cancer progression and to our ability to predict its risk [13, 23]. Targeted therapies incorporating inhibitors of the vascular endothelial growth factor (VEGF) pathway is the major treatment for patients with metastatic RCC [18, 20, 49]. For example, anti-angiogenic drugs such as sunitinib can nullify alterations in the microvascular properties of some human cancers [35]. This inhibition reduces tumor vascularization, and leads to tumor shrinkage. Clinically, cellular adaptations to VEGF-targeted anti-angiogenic therapy might induce drug resistance in RCC after transient disease stabilization, which limits the therapeutic efficacy of this approach and has been the major problem for this treatment. Therefore, a rational and efficacious treatment regimen for treating patients with RCC is needed [4, 14].

E2, is known to exert a protective effect beyond its classical endocrine role in several malignant diseases including neurodegenerative disorders, esophageal cancers, colorectal cancers, lung injuries, and coronary artery diseases [11, 40, 42, 46]. Many cellular functions can be improved by treatment with E2 alone or in combination with its receptors [3, 25]. For example, E2 can induce an increase in sensitivity to oxidative DNA damage through an ER-dependent pathway [28]. Moreover, the oxidative stress has been correlated to the phosphorylation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) at Ser40 [5, 29]. Therefore, the aim of this study was to understand whether E2 might induce different cellular effects on RCC cells. Briefly, changes in DNA repair mechanisms, oxidative stress response, and viability were determined to evaluate the altered cellular effects.

Methods

Cell culture and genetic characteristics of RCC cell lines

The incubation condition and clinical status of three RCC cell lines (ACHN, 786-O, and A498) were based on information disclosed on the website (https://www.atcc.org) of the American Type Culture Collection (Manassas, VA, USA). Their genetic characteristics are known from previous reports [9, 47]. In experiments that involved treatment with a specific chemical agent (or target hormone), subconfluent cell cultures were incubated with the indicated concentrations of E2 (E8875, Merck, Darmstadt, Germany) for the indicated times as our previous report [8]. Control cells were treated only with the solvent dimethyl sulfoxide (DMSO).

Immunodetection of estrogen receptors and Nrf2 from RCC cells

The protein levels of two estrogen receptors (ERs), ER1 and ER2, in RCC cells were determined by western blot analysis. For this, protein extracts of each RCC cell line were prepared with Pro-prep Protein Extraction Solution (iNtRON Biotechnology, Seongnam, South Korea) according to the manufacturer’s protocol with minor modifications. Briefly, the cell lysate was centrifuged for 10 min at 12,000 g to remove cellular debris. When analyzing the different protein fractions from the cytoplasm and nuclei, the different cellular compartments were extracted and separated using NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo Fisher Scientific, Rockford, IL, USA) according to the manufacturer’s instructions. Each protein concentration was determined using a BCA Protein Assay (Thermo Fisher Scientific). When performing western blotting, protein lysates were separated by sodium dodecyl sulfate polyacrylamide gel electrophoresis and transferred to polyvinylidene difluoride (PVDF) membranes. Then, each PVDF membrane was incubated with primary antibodies (anti-ER1, ab37438 and anti-ER2, ab3576; both from Abcam, Cambridge, MA, USA) for 1 h and then for another 1 h with horseradish peroxidase-labeled secondary antibody (L3012 for goat anti-rabbit IgG; Signalway Antibody, College Park, MD, USA) at room temperature. Target signals were exposed and enhanced using Western Lightning Plus-ECL Enhanced Luminol reagent (PK-NEL105, PerkinElmer, Waltham, MA, USA). In addition, the total Nrf2 and its phosphorylated form at Ser40 (phospho S40) in different cellular compartments were determined using specific antibody (ab62352 and ab76026, respectively; Abcam) from 10 μg aliquots of individual proteins. Target signals were amplified using the VECTASTAIN ABC-AmP kit (AK-6602, Vector Laboratories, Burlingame, CA, USA). The level of glyceraldehyde 3-phosphate dehydrogenase (anti-GAPDH antibody; AM4300, Life Technologies, Carlsbad, CA, USA) was determined as a loading control for whole-cell lysates. An anti-α-tubulin antibody (sc-5286, Santa Cruz Biochemicals, Dallas, TX, USA) was used as an internal control for cytoplasmic protein levels, and lamin A/C (anti-lamin A/C antibody; sc-7292, Santa Cruz Biochemicals) was used as an internal control for nuclear protein levels. Images of immunoblots were captured using a FluorChem FC2 system (Alpha Innotech, Santa Clara, CA, USA). Cell lysates of two breast cancer cell lines (T47-D and MCF-7) gifted from Prof. Shih-Ming Huang (Department and Graduate Institute of Biochemistry, National Defense Medical Center, Taiwan) served as positive controls for ER1 and ER2.

Assay for the formation of phosphorylated histone H2AX (γ-H2AX) foci

After incubation for 24 h of 5 × 104 cells per well, DNA double strand breaks (DSBs) were induced in A498 cells using etoposide as described in our previous report [17] and observed in the presence or absence of E2. The ability to repair DSBs was determined by staining with an anti-γ-H2AX antibody using an OxiSelect DNA Double Strand Break Staining Kit (Cell Biolabs, San Diego, CA, USA) according to the manufacturer’s instructions with some minor changes. Briefly, cells were allowed to recover by removing the DSB inducer for the indicated time; the exposed DSBs (shown as γ-H2AX foci) appeared as green fluorescence and nuclei were counterstained with 4′,6-diamidino-2-phenylindole. These fluorescent images were all detected using an Olympus IX70 fluorescence microscope (Olympus, Tokyo, Japan).

Detection of intracellular oxidative stress by flow cytometry

Intracellular oxidative stress induced by E2 treatment was determined by flow cytometry using 2′,7′-dichlorfluorescein-diacetate (DCFH-DA) as a sensitive nonfluorescent precursor dye according to a published standard procedure [1]. Cells were seeded on 6-well plates at 1.25 × 105 cells per well and cultured overnight as our previous report [8]. Then, cells were treated with E2 (7 or 28 μM) for 24 h and incubated with 50 μM DCFH-DA for 20 min in an incubator following an appropriate wash with phosphate-buffered saline (PBS). Cells were treated without E2 (DMSO alone) as a negative control or with 1 mM H2O2 for 30 min as a positive control. Cells were then washed again with PBS, incubated with 0.25 % trypsin- ethylenediaminetetraacetic acid for 1 min, and then quenched with minimum essential medium with 10 % fetal bovine serum to stop trypsinization. Detached cells were collected by centrifuging at 500 g for 5 min and resuspended in PBS. The fluorescence was determined by Flowcytometer FACSCalibur (BD Biosciences, Franklin Lakes, NJ, USA), with excitation at 480 nm and emission at 525 nm.

Detection of apoptosis

To identify apoptosis-positive cells, cells were stained with an Annexin V/propidium iodide (PI) double staining assay, with a fluorescein isothiocyanate (FITC) Annexin V Apoptosis Detection Kit I (BD Biosciences), according to the manufacturer’s protocol. Cells with various treatments were washed with buffer (20 mM Tris pH 7.4, 150 mM NaCl, 1 mM CaCl2) after incubation for 15 min in binding buffer containing 5 μL of Annexin V–FITC and 5 μL of PI at room temperature in the dark. Finally, fluorescent images of Annexin V–FITC binding and PI incorporation were detected using the previous fluorescence microscope.

Results

Diverse expression levels of ERs in different RCC cell lines

Three RCC cell lines (ACHN, 786-O, and A498) were cultured to detect the expression levels of ER1 and ER2 by western blot analysis (Fig. 1). Among these three cell lines, ACHN cells expressed the highest protein levels of ERs and A498 had the lowest expression, as reported previously for mRNA levels [8]. Therefore, these two RCC cell lines were subsequently analyzed for following cellular effects.

Fig. 1
figure 1

Immunodetection of estrogen receptors in cell lines of renal cell carcinoma by Western blots. Protein lysates were prepared from cell lines of renal cell carcinoma (ACHN, 786-O, and A498) and breast cancer (T47-D and MCF-7), which served as positive controls. GAPDH (glyceraldehyde-3-phosphate dehydrogenase) was the internal control. ER1 estrogen receptor 1, ER2 estrogen receptor 2

Induction of DNA repair by E2 in A498 cells

A498 cells (low expressions of ERs, wild-type p53, and VHL-null) had different properties of DNA repair under E2 treatment. As shown in Fig. 2, over 90 % (37 of 40) cells with active DNA repair were observed with green fluorescence for γH2AX under E2 treatment; whereas, less percentage of cells (44 %, 18 of 41) showed fluorescence-positive in the etoposide-treated group. This indicated that E2 could improve DNA repair, even though this RCC cell line had low expression levels of the ERs.

Fig. 2
figure 2

DNA repairing ability of A498 cells with treatments of etoposide and E2. A498 cells treated respectively with a etoposide (50 μM) alone or b etoposide (50 μM) and E2 (70 nM). Green, γH2AX-positive cells; blue, DAPI (4′,6-diamidino-2-phenylindole. The scale bar represented the 50 μm. E2, 17-β-estradiol

Induction of oxidative stress by E2 in ACHN cells

Reactive oxygen species (ROS), known to reflect oxidative stress, can cause significant damage to cell structures [33]. E2-induced ROS levels in ACHN cells (high ERs, wild-type p53, and VHL-positive) were detected using the DCFH-DA probe. As shown in Fig. 3, ACHN cells showed an increased trend in intracellular ROS levels when cells were treated with E2 (28 μM) for 24 h compared with the DMSO-treated negative control cells. In addition, activation of the Nrf2 transcription factor was observed (Fig. 4). Briefly, increased phosphorylation of nuclear Nrf2 (1.3 folds, in comparing to the cells treated with DMSO only) were detected in ACHN cells treated with E2 (28 μM), using immunoblot analysis.

Fig. 3
figure 3

Induction of oxidative stress by E2. Reactive oxidative species (ROS) formation in ACHN cells were detected by flow cytometry (28 μM E2, orange line). DCFH-DA-pretreated cells as ROS-positive were incubated with 50 μM DCFH-DA for 30 min at 37 °C. Blue line, 0.1 % DMSO (negative control); red line, 1 mM H2O2 (positive control). E2, 17-β-estradiol

Fig. 4
figure 4

Phosphorylation of nuclear Nrf2 by E2. Cytoplasm and nuclear extracts were respectively isolated from ACHN cells. Each compartment was immunodetected with antibody raised against phospho Nrf2 (Ser40), lamin A/C (for nuclear control), and α-tubulin (for cytosol control). Each protein level relative to individual control was determined by densitometry and shown on the bottom of each band. E2, 17-β-estradiol

Augmentation of apoptosis by E2 in ACHN cells

We next analyzed the apoptotic effects of E2 in ACHN cells by detecting Annexin V-positive cells. As shown in Fig. 5, ACHN cells treated with E2 (28 μM) could induce the higher binding rates (70 %, 7 of 10 E2 (10 nM)- and 75 %, 3 of 4 E2 (28 μM)-treated cells) of Annexin V to phosphatidyl serine at the surface of each cell. Conversely, only 10 % (2 of 20) cells shown Annexin V-positive in the DMSO-treated control group.

Fig. 5
figure 5

Apoptotic effect of E2 on ACHN cells. ACHN cells were treated without E2 (DMSO) and with E2 (10 nM and 28 μM). Annexin V, green; and PI (propidium iodide), red. E2, 17-β-estradiol. The scale bar represented the 50 μm

Discussion

Although the number of approved treatments for patients with advanced RCC continues to grow, additional work is still needed to delineate the optimal target drug or combination of agents for each subtype of this cancer [36]. The therapeutic management of RCC is mainly guided by the cancer stage and the degree of tumor dissemination, which affect the prognosis for patients directly [7]. Regardless of the clinic availability of the novel targeted therapeutics, and the favorable outcomes, however, complete or durable responses have been only rarely noted. Nearly all treated patients even now will develop drug resistance to any targeted treatment. Approximately 70 % of patients respond to therapy initially and the remaining 30 % show primary resistance (intrinsic resistance) [6, 15]. The 70 % of patients who show initial response, durable responses are rare, and acquired resistance (extrinsic resistance) to treatment develops in almost all of them in a median of 6–15 months [6, 15, 32]. However, it remains a challenge in the effective treatment of RCC and suggests that effective therapies for RCC need to be improved [12].

Autophagy mediated by E2 might have inhibited the growth rate of ACHN cells in the present study [8]. In addition, the activities of ERs have been correlated with those of VHL and p53 in many studies [2, 19, 41]. Therefore, the genetic status of VHL and p53 expressions of RCC cells should be considered when E2 is applied to treat RCC. p53-mediated transcription pathways are involved in ER-related pathways [26]. Loss of VHL may attenuate the DNA-damage response and result in the persistence of DSBs [27]. In the present results, A498 cells expressing wild-type p53 but with no VHL expression could repair etoposide-induced DNA damage in the presence of E2. The increased numbers of γH2AX-positive A498 cells implied that lower levels of ERs would not decrease their DNA repair capacity and that E2 treatment could facilitate repair of the DNA breaks even the cells were in the absence of VHL expression. This was similar to the conclusions of other authors who reported that complexes of E2 with ER1 or ER2 modulated the expression of many genes, including those involved in DNA repair [3, 22, 25, 39]. Moreover, the decreased DSBs would be obvious in the ACHN cells with wild type VHL expression. Here, we also found that ACHN cells expressed high levels of ERs. This might indicate that ACHN cells might be sensitive to E2 treatment and that this could induce many other cellular effects. This perspective also arose in other reports showing that E2 might be correlated to the development of this cancer [40, 45].

The highly conserved mechanism of autophagy is essential when cells are under stress, including that induced by ROS [44]. We reported previously that autophagosomes were observed in ACHN cells in the presence of E2 [8]. Sobočanec et al. and Zhu et al. recently reported that E2 could mediate some antioxidant enzymes via the Nrf2–Keap1 pathway [37, 48]. It will be agreed with our data which demonstrate an increased ROS under the E2 treatment. Here, we found that E2 could induce ROS production in ACHN cells. These E2-induced ROS might activate the Nrf2 pathway to reduce the oxidative stress and induce genotoxicity [30, 38]. Subsequently, sequential cell responses, autophagy, and apoptosis changed the fate of the ACHN cells. These findings were confirmed by the expression of activated nuclear Nrf2, a key transcription factor that regulates antioxidant defenses [21]. We found increased phosphorylation of Nrf2 in the nuclei of E2-treated ACHN cells, which might cause a delay in proliferation and induce apoptosis, similar to other cancers [34]. Taken together, our results indicate that Nrf2 activation may be necessary for this E2-induced autophagy and apoptosis. Combination therapy with E2 and target therapeutic agents may be a safe and potential practice clinically to improve the tolerance and treatment duration of target therapy.

Conclusions

Adjuvant therapy with sex hormones could be beneficial in many human diseases [8, 24]. Here, we demonstrated the cellular effects of E2 on DNA repair, ROS production as well as Nrf2 activation, and apoptosis in RCC cell lines. It implies that these cellular alterations may contribute to the reduced viability of RCC cells following E2 treatment.

Abbreviations

DCFH-DA:

2′,7′-dichlorfluorescein-diacetate

E2:

17-β-estradiol

ER1:

E2 receptor 1

ER2:

E2 receptor 2

Nrf2:

Nuclear factor (erythroid-derived 2)-like 2

RCC:

Renal cell carcinoma

ROS:

Reactive oxygen species

VEGF:

Vascular endothelial growth factor

References

  1. Ashida S, Nishimori I, Tanimura M, Onishi S, Shuin T. Effects of von Hippel-Lindau gene mutation and methylation status on expression of transmembrane carbonic anhydrases in renal cell carcinoma. J Cancer Res Clin Oncol. 2002;128(10):561–8.

    Article  CAS  PubMed  Google Scholar 

  2. Bado I, Nikolos F, Rajapaksa G, Gustafsson JA, Thomas C. ERbeta decreases the invasiveness of triple-negative breast cancer cells by regulating mutant p53 oncogenic function. Oncotarget. 2016;7(12):13599–611.

    PubMed  PubMed Central  Google Scholar 

  3. Boland R, Vasconsuelo A, Milanesi L, Ronda AC, de Boland AR. 17beta-estradiol signaling in skeletal muscle cells and its relationship to apoptosis. Steroids. 2008;73(9–10):859–63.

    Article  CAS  PubMed  Google Scholar 

  4. Bridgeman VL, Wan E, Foo S, Nathan MR, Welti JC, Frentzas S, Vermeulen PB, Preece N, Springer CJ, Powles T, Nathan PD, Larkin J, Gore M, Vasudev NS, Reynolds AR. Preclinical evidence that trametinib enhances the response to antiangiogenic tyrosine kinase inhibitors in renal cell carcinoma. Mol Cancer Ther. 2016;15(1):172–83.

    Article  CAS  PubMed  Google Scholar 

  5. Buelna-Chontal M, Guevara-Chavez JG, Silva-Palacios A, Medina-Campos ON, Pedraza-Chaverri J, Zazueta C. Nrf2-regulated antioxidant response is activated by protein kinase C in postconditioned rat hearts. Free Radic Biol Med. 2014;74:145–56.

    Article  CAS  PubMed  Google Scholar 

  6. Busch J, Seidel C, Weikert S, Wolff I, Kempkensteffen C, Weinkauf L, Hinz S, Magheli A, Miller K, Grunwald V. Intrinsic resistance to tyrosine kinase inhibitors is associated with poor clinical outcome in metastatic renal cell carcinoma. BMC Cancer. 2011;11:295.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Chen DY, Uzzo RG. Optimal management of localized renal cell carcinoma: surgery, ablation, or active surveillance. J Natl Compr Canc Netw. 2009;7(6):635–42. quiz 643.

    PubMed  PubMed Central  Google Scholar 

  8. Chen KC, Lin CM, Huang CJ, Chen SK, Wu ST, Chiang HS, Ku WC. Dual roles of 17-beta estradiol in estrogen receptor-dependent growth inhibition in renal cell carcinoma. Cancer Genomics Proteomics. 2016;13(3):219–30.

    CAS  PubMed  Google Scholar 

  9. Cho DC, Cohen MB, Panka DJ, Collins M, Ghebremichael M, Atkins MB, Signoretti S, Mier JW. The efficacy of the novel dual PI3-kinase/mTOR inhibitor NVP-BEZ235 compared with rapamycin in renal cell carcinoma. Clin Cancer Res. 2010;16(14):3628–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Czarnecka AM, Niedzwiedzka M, Porta C, Szczylik C. Hormone signaling pathways as treatment targets in renal cell cancer (Review). Int J Oncol. 2016;48(6):2221–35.

    PubMed  Google Scholar 

  11. D’Alessandro A, D’Aguanno S, Cencioni MT, Pieroni L, Diamantini A, Battistini L, Longone P, Spalloni A, De Laurenzi V, Bernardini S, Federici G, Urbani A. Protein repertoire impact of Ubiquitin-Proteasome System impairment: insight into the protective role of beta-estradiol. J Proteomics. 2012;75(4):1440–53.

    Article  PubMed  Google Scholar 

  12. Farber LJ, Furge K, Teh BT. Renal cell carcinoma deep sequencing: recent developments. Curr Oncol Rep. 2012;14(3):240–8.

    Article  CAS  PubMed  Google Scholar 

  13. Feng C, Xiong Z, Jiang H, Ding Q, Fang Z, Hui W. Genetic alteration in notch pathway is associated with better prognosis in renal cell carcinoma. BioFactors. 2016;42(1):41–8.

    CAS  PubMed  Google Scholar 

  14. Han KS, Raven PA, Frees S, Gust K, Fazli L, Ettinger S, Hong SJ, Kollmannsberger C, Gleave ME, So AI. Cellular adaptation to VEGF-targeted antiangiogenic therapy induces evasive resistance by overproduction of alternative endothelial cell growth factors in renal cell carcinoma. Neoplasia. 2015;17(11):805–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Heng DY, Mackenzie MJ, Vaishampayan UN, Bjarnason GA, Knox JJ, Tan MH, Wood L, Wang Y, Kollmannsberger C, North S, Donskov F, Rini BI, Choueiri TK. Primary anti-vascular endothelial growth factor (VEGF)-refractory metastatic renal cell carcinoma: clinical characteristics, risk factors, and subsequent therapy. Ann Oncol. 2012;23(6):1549–55.

    Article  CAS  PubMed  Google Scholar 

  16. Hew MN, Zonneveld R, Kummerlin IP, Opondo D, de la Rosette JJ, Laguna MP. Age and gender related differences in renal cell carcinoma in a European cohort. J Urol. 2012;188(1):33–8.

    Article  PubMed  Google Scholar 

  17. Huang CJ, Yang SH, Lee CL, Cheng YC, Tai SY, Chien CC. Ribosomal protein S27-like in colorectal cancer: a candidate for predicting prognoses. PLoS One. 2013;8(6):e67043.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic therapy in oncology: current status and future directions. Lancet. 2016; 388(10043):518–29.

    Article  CAS  PubMed  Google Scholar 

  19. Jung YS, Chun HY, Yoon MH, Park BJ. Elevated estrogen receptor-alpha in VHL-deficient condition induces microtubule organizing center amplification via disruption of BRCA1/Rad51 interaction. Neoplasia. 2014;16(12):1070–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kaymakcalan MD, Xie W, Albiges L, North SA, Kollmannsberger CK, Smoragiewicz M, Kroeger N, Wells JC, Rha SY, Lee JL, McKay RR, Fay AP, De Velasco G, Heng DY, Choueiri TK. Risk factors and model for predicting toxicity-related treatment discontinuation in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy: results from the International Metastatic Renal Cell Carcinoma Database Consortium. Cancer. 2016;122(3):411–9.

    Article  CAS  PubMed  Google Scholar 

  21. Li N, Alam J, Venkatesan MI, Eiguren-Fernandez A, Schmitz D, Di Stefano E, Slaughter N, Killeen E, Wang X, Huang A. Nrf2 is a key transcription factor that regulates antioxidant defense in macrophages and epithelial cells: protecting against the proinflammatory and oxidizing effects of diesel exhaust chemicals. J Immunol. 2004;173(5):3467–81.

    Article  CAS  PubMed  Google Scholar 

  22. Lin CY, Strom A, Vega VB, Kong SL, Yeo AL, Thomsen JS, Chan WC, Doray B, Bangarusamy DK, Ramasamy A, Vergara LA, Tang S, Chong A, Bajic VB, Miller LD, Gustafsson JA, Liu ET. Discovery of estrogen receptor alpha target genes and response elements in breast tumor cells. Genome Biol. 2004;5(9):R66.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Loginov VI, Dmitriev AA, Senchenko VN, Pronina IV, Khodyrev DS, Kudryavtseva AV, Krasnov GS, Gerashchenko GV, Chashchina LI, Kazubskaya TP, Kondratieva TT, Lerman MI, Angeloni D, Braga EA, Kashuba VI. Tumor suppressor function of the SEMA3B gene in human lung and renal cancers. PLoS One. 2015;10(5):e0123369.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Masood DE, Roach EC, Beauregard KG, Khalil RA. Impact of sex hormone metabolism on the vascular effects of menopausal hormone therapy in cardiovascular disease. Curr Drug Metab. 2010;11(8):693–714.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Mateos L, Persson T, Kathozi S, Gil-Bea FJ, Cedazo-Minguez A. Estrogen protects against amyloid-β toxicity by estrogen receptor α-mediated inhibition of Daxx translocation. Neurosci Lett. 2012;506(2):245–50.

    Article  CAS  PubMed  Google Scholar 

  26. Menendez D, Inga A, Resnick MA. Estrogen receptor acting in cis enhances WT and mutant p53 transactivation at canonical and noncanonical p53 target sequences. Proc Natl Acad Sci. 2010;107(4):1500–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Metcalf JL, Bradshaw PS, Komosa M, Greer SN, Stephen Meyn M, Ohh M. K63-ubiquitylation of VHL by SOCS1 mediates DNA double-strand break repair. Oncogene. 2014;33(8):1055–65.

    Article  CAS  PubMed  Google Scholar 

  28. Mobley JA, Brueggemeier RW. Estrogen receptor-mediated regulation of oxidative stress and DNA damage in breast cancer. Carcinogenesis. 2004;25(1):3–9.

    Article  CAS  PubMed  Google Scholar 

  29. Mota SI, Costa RO, Ferreira IL, Santana I, Caldeira GL, Padovano C, Fonseca AC, Baldeiras I, Cunha C, Letra L, Oliveira CR, Pereira CM, Rego AC. Oxidative stress involving changes in Nrf2 and ER stress in early stages of Alzheimer’s disease. Biochim Biophys Acta. 2015;1852(7):1428–41.

    Article  CAS  PubMed  Google Scholar 

  30. Okoh V, Deoraj A, Roy D. Estrogen-induced reactive oxygen species-mediated signalings contribute to breast cancer. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2011;1815(1):115–33.

    Article  CAS  Google Scholar 

  31. Rampersaud EN, Klatte T, Bass G, Patard JJ, Bensaleh K, Bohm M, Allhoff EP, Cindolo L, De La Taille A, Mejean A, Soulie M, Bellec L, Christophe Bernhard J, Pfister C, Colombel M, Belldegrun AS, Pantuck AJ, George D. The effect of gender and age on kidney cancer survival: younger age is an independent prognostic factor in women with renal cell carcinoma. Urol Oncol. 2014;32(1):30.e9–13.

  32. Rini BI, Atkins MB. Resistance to targeted therapy in renal-cell carcinoma. The Lancet Oncology. 2009;10(10):992–1000.

    Article  CAS  PubMed  Google Scholar 

  33. Roy D, Cai Q, Felty Q, Narayan S. Estrogen-induced generation of reactive oxygen and nitrogen species, gene damage, and estrogen-dependent cancers. J Toxicol Environ Health B Crit Rev. 2007;10(4):235–57.

    Article  CAS  PubMed  Google Scholar 

  34. Schafer G, H Kaschula C. The immunomodulation and anti-inflammatory effects of garlic organosulfur compounds in cancer chemoprevention. Anticancer Agents Med Chem. 2014;14(2):233–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Schroeder M, Viezens L, Wellbrock J, Fiedler W, Ruther W, Algenstaedt P, Hansen-Algenstaedt N, Schaefer C. Sunitinib treatment reduces tumor growth and limits changes in microvascular properties after minor surgical intervention in an in vivo model of secondary breast cancer growth in bone. J Surg Oncol. 2016;113(5):515–21.

    Article  CAS  PubMed  Google Scholar 

  36. Singer EA, Gupta GN, Marchalik D, Srinivasan R. Evolving therapeutic targets in renal cell carcinoma. Curr Opin Oncol. 2013;25(3):273–80.

    CAS  PubMed  Google Scholar 

  37. Sobocanec S, Saric A, Macak Safranko Z, Popovic Hadzija M, Abramic M, Balog T. The role of 17beta-estradiol in the regulation of antioxidant enzymes via the Nrf2-Keap1 pathway in the livers of CBA/H mice. Life Sci. 2015;130:57–65.

    Article  CAS  PubMed  Google Scholar 

  38. To C, Ringelberg CS, Royce DB, Williams CR, Risingsong R, Sporn MB, Liby KT. Dimethyl fumarate and the oleanane triterpenoids, CDDO-imidazolide and CDDO-methyl ester, both activate the Nrf2 pathway but have opposite effects in the A/J model of lung carcinogenesis. Carcinogenesis. 2015;36(7):769–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Vasconsuelo A, Milanesi L, Boland R. 17Beta-estradiol abrogates apoptosis in murine skeletal muscle cells through estrogen receptors: role of the phosphatidylinositol 3-kinase/Akt pathway. J Endocrinol. 2008;196(2):385–97.

    Article  CAS  PubMed  Google Scholar 

  40. Wang QM, Qi YJ, Jiang Q, Ma YF, Wang LD. Relevance of serum estradiol and estrogen receptor beta expression from a high-incidence area for esophageal squamous cell carcinoma in China. Med Oncol. 2011;28(1):188–93.

    Article  CAS  PubMed  Google Scholar 

  41. Wang X, Simpson ER, Brown KA. p53: protection against tumor growth beyond effects on cell cycle and apoptosis. Cancer Res. 2015;75(23):5001–7.

    Article  CAS  PubMed  Google Scholar 

  42. Weige CC, Allred KF, Allred CD. Estradiol alters cell growth in nonmalignant colonocytes and reduces the formation of preneoplastic lesions in the colon. Cancer Res. 2009;69(23):9118–24.

    Article  CAS  PubMed  Google Scholar 

  43. Weng WH, Chen YT, Yu KJ, Chang YH, Chuang CK, Pang ST. Genetic alterations of HER genes in chromophobe renal cell carcinoma. Oncol Lett. 2016;11(3):2111–6.

    PubMed  PubMed Central  Google Scholar 

  44. Wu JJ, Quijano C, Chen E, Liu H, Cao L, Fergusson MM, Rovira II, Gutkind S, Daniels MP, Komatsu M, Finkel T. Mitochondrial dysfunction and oxidative stress mediate the physiological impairment induced by the disruption of autophagy. Aging (Albany NY). 2009;1(4):425–37.

    Article  CAS  Google Scholar 

  45. Yu CP, Ho JY, Huang YT, Cha TL, Sun GH, Yu DS, Chang FW, Chen SP, Hsu RJ. Estrogen inhibits renal cell carcinoma cell progression through estrogen receptor-beta activation. PLoS One. 2013;8(2):e56667.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Yu GS, Lai CY, Xu Y, Bu CF, Su ZX. Aberrant methylation of RASSF1A gene contribute to the risk of renal cell carcinoma: a meta-analysis. Asian Pac J Cancer Prev. 2015;16(11):4665–9.

    Article  PubMed  Google Scholar 

  47. Yuan J, Luo K, Zhang L, Cheville JC, Lou Z. USP10 regulates p53 localization and stability by deubiquitinating p53. Cell. 2010;140(3):384–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhu C, Wang S, Wang B, Du F, Hu C, Li H, Feng Y, Zhu R, Mo M, Cao Y, Li A, Yu X. 17beta-Estradiol up-regulates Nrf2 via PI3K/AKT and estrogen receptor signaling pathways to suppress light-induced degeneration in rat retina. Neuroscience. 2015;304:328–39.

    Article  CAS  PubMed  Google Scholar 

  49. Zisman A, Patard JJ, Raz O, Klatte T, Haifler M, Mendlovic S, Sandbank J, Belldegrun AS, Lindner A, Leibovici D, Pantuck AJ. Sex, age, and surgeon decision on nephron-sparing surgery are independent predictors of renal masses with benign histologic findings–a multicenter survey. Urology. 2010;76(3):541–6.

    Article  PubMed  Google Scholar 

Download references

Acknowledgments

The authors thank Ms. Hsiao-Ting Tseng for her technical support.

Funding

The work was supported by grants from Cathay General Hospital (CGH-MR-10203 to SKC and 102-CGH-FJU-18 to WCK), Fu Jen Catholic University (A0104028 to WCK), and Ministry of Science and Technology (NSC 102-2320-B-030-012-MY2 to WCK).

Availability of data and materials

https://drive.google.com/open?id=0B1QtbaZpq7Fqb3dGd1NOVTdvNUU.

Authors’ contributions

STW, CJH, WCK and SKC designed research. STW, CJH, and SKC performed research. CJH, YCW, and CML analyzed data. STW, WCK, CJH, and SKC wrote paper. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Shao-Kuan Chen.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, ST., Ku, WC., Huang, CJ. et al. Cellular effects induced by 17-β-estradiol to reduce the survival of renal cell carcinoma cells. J Biomed Sci 23, 67 (2016). https://doi.org/10.1186/s12929-016-0282-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12929-016-0282-z

Keywords