Skip to main content
Erschienen in: Journal of Ovarian Research 1/2013

Open Access 01.12.2013 | Research

Sulforaphane induced cell cycle arrest in the G2/M phase via the blockade of cyclin B1/CDC2 in human ovarian cancer cells

verfasst von: Chi-Chang Chang, Chao-Ming Hung, Yun-Ru Yang, Mon-Juan Lee, Yi-Chiang Hsu

Erschienen in: Journal of Ovarian Research | Ausgabe 1/2013

Abstract

Background

Malignant tumors are the single most common cause of death and the mortality rate of ovarian cancer is the highest among gynecological disorders. The excision of benign tumors is generally followed by complete recovery; however, the activity of cancer cells often results in rapid proliferation even after the tumor has been excised completely. Thus, clinical treatment must be supplemented by auxiliary chemotherapy or radiotherapy. Sulforaphane (SFN) is an extract from the mustard family recognized for its anti-oxidation abilities, phase 2 enzyme induction, and anti-tumor activity.

Methods

This study investigated the cell cycle arrest in G2/M by SFN and the expression of cyclin B1, Cdc2, and the cyclin B1/CDC2 complex in PA-1 cells using western blotting and co-IP western blotting.

Results

This study investigated the anticancer effects of dietary isothiocyanate SFN on ovarian cancer, using cancer cells line PA-1. SFN-treated cells accumulated in metaphase by CDC2 down-regulation and dissociation of the cyclin B1/CDC2 complex.

Conclusion

Our findings suggest that, in addition to the known effects on cancer prevention, SFN may also provide antitumor activity in established ovarian cancer.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1757-2215-6-41) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

CCC participated in its design and coordination and drafted the manuscript. YRY carried out the FACS analysis, cell culture and carried out the molecular studies. CMH performed the statistical analysis. YCH conceived of the study, and participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.

Background

Isothiocyanates (ITCs) are naturally occurring components of vegetables that have demonstrated biological activity against carcinogenesis as well as chemopreventive properties [1]. It has been suggested that in conjunction with chemotherapy, ITCs may enhance drug sensitivity [2]. Sulforaphane (SFN), a potent cancer preventive agent, is a dietary isothiocyanate found as a precursor glucosinolate in cruciferous vegetables such as Brussels sprouts, cauliflower and broccoli [3]. Interest in this agent has grown in recent years due to its putative beneficial pharmacological effects as an antioxidant [4], anti-inflammatory [5] and antitumor agent [6]. SFN is also a potent scavenger of reactive oxygen species (ROS), including superoxide anions and hydroxyl radicals [7]. Many studies have indicated an inverse correlation between the consumption of cruciferous vegetables and a decrease in the incidence of various tumors, including those of the prostate [8], cervical [9], colorectal [10], and lung [11]. In addition to inhibiting cell proliferation and increasing apoptosis [10], other mechanisms have also been proposed to explain the anti-carcinogenic effects of SFN. These include anti-inflammatory and antioxidative activities, the induction of phase 2 detoxification enzymes, the inhibition of cyclooxygenase 2 (COX-2) [12], and the effect on protein kinases [13].
This study investigated the influence of SFN on ovarian cancer cell lines (PA-1) with regard to the anti-proliferation of PA-1 cells and induced cell cycle arrest in the G2/M phase. These results may provide support for the chemoprevention of ovarian cancer.

Methods

Materials

Sulforaphane [1-isothiocyanato-(4R,S)-(methylsulfinyl)butane], DMSO (dimethyl sulfoxide) and MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide], were obtained from Sigma (St Louis, MO). All other reagents and compounds were analytical grade.

Cell culture

PA-1 cells were purchased from the Food Industry Research and Development Institute (Hsinchu, Taiwan). The cells were maintained in flasks containing MEM supplemented with 10% (v/v) FBS and cultured in an incubator at 37°C with an atmosphere containing 5% CO2.

Cell proliferation assay

Cells were seeded into 96-well culture plates at 10,000 cells/well and treated with SFN. One to three days (0 μM SFN was the control group.) MTT dye (1 mg/ml) was added to each well 4 hours following treatment. The reaction was stopped by the addition of DMSO, and OD540 was measured using a multi-well plate reader (Powerwave XS, Biotek). In the absence of cells, the background absorbance of the medium was subtracted. Results were expressed as a percentage of the control, which was considered to be 100%. Each assay was performed in triplicate and the results were expressed as the mean (+/−SEM).

Measurement of apoptosis

PA-1 cells were first seeded in 6-well plates (Orange Scientific, E.U.). Following treatment with SFN for four hours, the supernatant was discarded and cells were harvested and re-centrifuged. Cells were subsequently resuspended/incubated in 1X annexin-binding buffer (5 μL of annexin V-FITC [BD Pharmingen, BD, USA] and 1 μL of 100 μg/mL PI working solution) for 15 minutes. Following incubation, the stained cells were analyzed using flow cytometry (FACSCalibur, BD, USA). Data was analyzed using WinMDI 2.8 free software (BD, USA).

Cell cycle analysis

Cell cycle analysis was performed using fluorescent nucleic acid dye and propidium iodide (PI) to identify the proportion of cells in each of the three stages of interphase. Cells were treated with SFN for 24 hours, and subsequently harvested and fixed in 1 ml of cold 70% ethanol for at least eight hours at −20°C. DNA was stained in PI/RNaseA solution and the DNA content was detected using flow cytometry. Data was analyzed using WinMDI 2.8 free software (BD, USA).

Western blot assay

A total of 30–50 μg of proteins were separated by 10% SDS-PAGE and transferred to PVDF membranes (Millipore, USA). The membranes were blocked with blocking buffer (Odyddey, USA) overnight and subsequently incubated with anti-β-actin (Sigma-Aldrich, St. Louis, MO, USA), anti-CDC2, anti-caspase 3, and anti-cyclin B1 (Santa Cruz BioTechnology, USA) antibodies for 1.5 ~ 2 hours. The blots were then washed and incubated with a second antibody (IRDye Li-COR, USA) at a dilution of 1/20,000 for 30 minutes. Finally, the antigen was visualized using a near infrared imaging system (Odyssey LI-COR, USA) and data was analyzed using Odyssey 2.1 software.

Co-immunoprecipitation (Co-IP)

Co-IP is an effective means of quantifying protein-protein interaction in cells. Briefly, 500 μg of cellular proteins were labeled using anti-cyclin B1 (Santa Cruz BioTechnology, USA) following overnight incubation at room temperature. The protein-antibody immunoprecipitates were collected by protein A/G plus-agarose (Santa Cruz BioTechnology, USA). Following the final wash, the samples were boiled and centrifuged to pellet the agarose beads. Western blot analysis of the CDC2 protein in the supernatant was then conducted. Antigens were visualized using a near infrared imaging system (Odyssey LI-COR, USA) and data was analyzed using Odyssey 2.1 software.

Statistical analysis

All data was reported as the mean (±SEM) of at least three separate experiments. A t-test or one-way ANOVA with post-hoc test was employed for statistical analysis, with significant differences determined as P < 0.05.

Results

SFN inhibits proliferation of PA-1 cells

We hypothesized that SFN could mediate the survival of PA-1 cells and thus inhibit their growth. To explore the anti-tumor activity of SFN against PA-1 cells, an in vitro cell viability study was conducted in which each sample of the PA-1 cells was treated with increasing doses of SFN (0, 6.25, and 12.5 μM) for 24 to 72 hours. The results in (Figure 1A) indicate that the survival and proliferation of PA-1 cells were decreased by treatment with SFN in a dose-dependent (The 24 hours’ IC50 of simvastatin in the PA-1 cancer cells was determined to be 17.083 μM; y = −2.8213× + 98.199, R2 = 0.9697) (*p < 0.05 vs SFN 0 μM group) and time-course manner (& p < 0.05 vs 24 hour treatment, # p < 0.05 vs 48 hour treatment).

SFN repressed the cell viability of PA-1 cells without apoptosis induction

A study on apoptosis was performed to further elucidate anti-cancer mechanisms of SFN in PA-1 cells. After treating the cells with various doses of SFN, the percentage of apoptotic cells was assessed using Annexin V-FITC and propidium iodide staining, followed by flow cytometric analysis (Figure 1B). A dot-plot of Annexin V-FITC fluorescence versus PI fluorescence also indicated a non-significant increase in the percentage of apoptotic cells treated with SFN. At SFN concentrations of 6.25 to 12.5 μM, no significant increase was observed in the percentage of cells undergoing necrosis and apoptosis (Figure 1C) or caspase 3 activation (Figure 1D). The results summarized in Figure 1 indicate that SFN may mediate the survival of PA-1 cells and thus inhibit their proliferation without apoptosis induction.

SFN treatment induced the accumulation of G2/M phase in PA-1 cells

The cell-cycle distribution of SFN-treated PA-1 cells was analyzed using flow cytometry. Prior to processing and analysis, cells were exposed to SFN for a total of 24 hours. As shown in Figure 2A, the cells exposed to SFN showed an increase in the number of cells in the G2/M phase (& p < 0.05 vs SFN 0 μM), compared with the number of untreated cells. This could imply that the PA-1 cells had undergone cell cycle arrest. We also found that treatment with SFN simultaneously reduced the number of cells in the G1 phase (* p < 0.05 vs SFN 0 μM) (Figure 2B).

Cell cycle arrest by SFN in PA-1 cells via CDC2 down regulation and dissociation of the cyclin B1/CDC2 complex

Figure 3 illustrates the immunoblotting of cellular proteins from PA-1 cells treated with SFN, revealing a decrease in CDC2 following incubation with SFN (Figure 3A). Cyclin B1 and CDC2 protein expression was quantified by measuring relative intensities. We found that CDC2 levels were significantly lower in cells incubated with SFN concentrations of 12.5 μM (Figure 3B). Moreover, the activity of the cyclin B1/CDC2 complex (important for G2-M transition during the cell cycle) was determined by Co-IP (Figure 4A) and quantified by measuring relative band intensities. We found that cyclin B1/CDC2 complex activity was significantly suppressed in cells incubated with an SFN concentration of 12.5 μM (Figure 4B).
These results indicate an increase of the cell population in G2/M phase via a down regulation of CDC2 and dissociation of the cyclin B1/CDC2 complex following incubation with SFN in PA-1 cells.

Discussion

The results collected in this study using cell lines of human ovarian cancer provide experimental evidence indicating that SFN may induce irreversible cell cycle arrest during the G2/M phase. These dietary constituents demonstrate chemopreventive and chemotherapeutic potential through their ability to ameliorate the side effects of conventional chemotherapy [14].
This study investigated the in vitro expression of cyclin B1 and Cdc2 in PA-1 cells. The cells possess mechanisms to maintain genomic stability through cell cycle arrest [15]. At least two cell cycle checkpoints play a role in the cellular response, allowing the DNA to be repaired prior to DNA duplication (G1/S checkpoint) or mitosis (G2/M checkpoint) [16]. Cdc2 regulates mitosis and binds to cyclin B to form mitosis-promoting factor (MPF) [17]. The activity of MPF is regulated by the phosphorylation/dephosphorylation of Cdc2 as well as the accumulation of cyclin B protein and p53; GADD45 is also involved in a G2/M checkpoint and may participate in the regulation of Cdc2 kinase activity [18, 19].
Other lines of evidence suggest that DNA damage excludes cyclin B1 from the nucleus, which promotes G2 arrest [20]. It is possible that cell cycle checkpoints delay cell cycle progression to allow additional time for the repair of DNA damage; however, our study found no direct evidence that DNA was repaired while the cells were arrested at the checkpoint.
Recent studies have shown that SFN inhibits the growth of tumor precursors and tumors in mice models when treatment is initiated at the time of carcinogen administration [10]. The co-inhibition of PI3K/AKT and ERK pathways activates FOXO transcription factor and enhances SFN-induced FOXO transcriptional activity, leading to cell cycle arrest and apoptosis [21].

Conclusions

We conclude that G2 delay is a common response of tumor cells to chemotherapy with SFN. We further propose that mechanisms of this delay may be reduced expression of CDC2 and dissociation of the cyclin B1/CDC2 complex. Therefore, although certain chemopreventive effects of SFN and related isothiocyanate compounds have already been established with regard to ovarian cancer cells, SFN should be investigated further to confirm the additional antitumor properties proposed by our study.

Acknowledgment

The authors would like to express their appreciation for the funding support provided by the E-Da Hospital, E-Da Hospital/I-Shou University. (EDAHP-100018).
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

CCC participated in its design and coordination and drafted the manuscript. YRY carried out the FACS analysis, cell culture and carried out the molecular studies. CMH performed the statistical analysis. YCH conceived of the study, and participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Singh SV, Singh K: Cancer chemoprevention with dietary isothiocyanates mature for clinical translational research. Carcinogenesis 2012, 33: 1833–1842. 10.1093/carcin/bgs216PubMedCentralPubMedCrossRef Singh SV, Singh K: Cancer chemoprevention with dietary isothiocyanates mature for clinical translational research. Carcinogenesis 2012, 33: 1833–1842. 10.1093/carcin/bgs216PubMedCentralPubMedCrossRef
2.
Zurück zum Zitat Xu T, Ren D, Sun X, Yang G: Dual Roles of Sulforaphane in Cancer Treatment. Anticancer Agents Med Chem 2012, 12: 1132–1142. 10.2174/187152012803529691PubMedCrossRef Xu T, Ren D, Sun X, Yang G: Dual Roles of Sulforaphane in Cancer Treatment. Anticancer Agents Med Chem 2012, 12: 1132–1142. 10.2174/187152012803529691PubMedCrossRef
3.
Zurück zum Zitat Huang TY, Chang WC, Wang MY, Yang YR, Hsu YC: Effect of sulforaphane on growth inhibition in human brain malignant glioma GBM 8401 cells by means of mitochondrial- and MEK/ERK-mediated apoptosis pathway. Cell Biochem Biophys 2012, 63: 247–259. 10.1007/s12013-012-9360-3PubMedCrossRef Huang TY, Chang WC, Wang MY, Yang YR, Hsu YC: Effect of sulforaphane on growth inhibition in human brain malignant glioma GBM 8401 cells by means of mitochondrial- and MEK/ERK-mediated apoptosis pathway. Cell Biochem Biophys 2012, 63: 247–259. 10.1007/s12013-012-9360-3PubMedCrossRef
4.
Zurück zum Zitat Alp H, Aytekin I, Hatipoglu NK, Alp A, Ogun M: Effects of sulforophane and curcumin on oxidative stress created by acute malathion toxicity in rats. Eur Rev Med Pharmacol Sci 2012, 16: 144–148.PubMed Alp H, Aytekin I, Hatipoglu NK, Alp A, Ogun M: Effects of sulforophane and curcumin on oxidative stress created by acute malathion toxicity in rats. Eur Rev Med Pharmacol Sci 2012, 16: 144–148.PubMed
5.
Zurück zum Zitat Yanaka A, Sato J, Ohmori S: Sulforaphane protects small intestinal mucosa from aspirin/NSAID-induced injury by enhancing host defense systems against oxidative stress and by inhibiting mucosal invasion of anaerobic enterobacteria. Curr Pharm Des 2013, 19: 157–162.PubMed Yanaka A, Sato J, Ohmori S: Sulforaphane protects small intestinal mucosa from aspirin/NSAID-induced injury by enhancing host defense systems against oxidative stress and by inhibiting mucosal invasion of anaerobic enterobacteria. Curr Pharm Des 2013, 19: 157–162.PubMed
6.
Zurück zum Zitat Devi JR, Thangam EB: Mechanisms of anticancer activity of sulforaphane from Brassica oleracea in HEp-2 human epithelial carcinoma cell line. Asian Pac J Cancer Prev 2012, 13: 2095–2100. 10.7314/APJCP.2012.13.5.2095PubMedCrossRef Devi JR, Thangam EB: Mechanisms of anticancer activity of sulforaphane from Brassica oleracea in HEp-2 human epithelial carcinoma cell line. Asian Pac J Cancer Prev 2012, 13: 2095–2100. 10.7314/APJCP.2012.13.5.2095PubMedCrossRef
7.
Zurück zum Zitat Lin LC, Yeh CT, Kuo CC, Lee CM, Yen GC, Wang LS, Wu CH, Yang WC, Wu AT: Sulforaphane potentiates the efficacy of imatinib against chronic leukemia cancer stem cells through enhanced abrogation of Wnt/β-catenin function. J Agric Food Chem 2012, 60: 7031–7039. 10.1021/jf301981nPubMedCrossRef Lin LC, Yeh CT, Kuo CC, Lee CM, Yen GC, Wang LS, Wu CH, Yang WC, Wu AT: Sulforaphane potentiates the efficacy of imatinib against chronic leukemia cancer stem cells through enhanced abrogation of Wnt/β-catenin function. J Agric Food Chem 2012, 60: 7031–7039. 10.1021/jf301981nPubMedCrossRef
8.
Zurück zum Zitat Wiczk A, Hofman D, Konopa G, Herman-Antosiewicz A: Sulforaphane, a cruciferous vegetable-derived isothiocyanate, inhibits protein synthesis in humanprostate cancer cells. Biochim Biophys Acta 1823, 2012: 1295–1305. Wiczk A, Hofman D, Konopa G, Herman-Antosiewicz A: Sulforaphane, a cruciferous vegetable-derived isothiocyanate, inhibits protein synthesis in humanprostate cancer cells. Biochim Biophys Acta 1823, 2012: 1295–1305.
9.
Zurück zum Zitat Hussain A, Priyani A, Sadrieh L, Brahmbhatt K, Ahmed M, Sharma C: Concurrent sulforaphane and eugenol induces differential effects on human cervical cancer cells. Integr Cancer Ther 2012, 11: 154–165.PubMedCrossRef Hussain A, Priyani A, Sadrieh L, Brahmbhatt K, Ahmed M, Sharma C: Concurrent sulforaphane and eugenol induces differential effects on human cervical cancer cells. Integr Cancer Ther 2012, 11: 154–165.PubMedCrossRef
10.
Zurück zum Zitat Chen MJ, Tang WY, Hsu CW, Wu JF, Lin CW, Cheng YM, Hsu YC: Apoptosis induction in primary human colorectal cancer cell lines and retarded tumor growth in SCID mice by sulforaphane. Evid Based Complement Alternat Med 2012,201(2):415231. Chen MJ, Tang WY, Hsu CW, Wu JF, Lin CW, Cheng YM, Hsu YC: Apoptosis induction in primary human colorectal cancer cell lines and retarded tumor growth in SCID mice by sulforaphane. Evid Based Complement Alternat Med 2012,201(2):415231.
11.
Zurück zum Zitat Kombairaju P, Ma J, Thimmulappa RK, Yan SG, Gabrielson E, Singh A, Biswal S: Prolonged sulforaphane treatment does not enhance tumorigenesis in oncogenic K-ras and xenograft mouse models of lung cancer. J Carcinog 2012, 11: 8. 10.4103/1477-3163.98459PubMedCentralPubMedCrossRef Kombairaju P, Ma J, Thimmulappa RK, Yan SG, Gabrielson E, Singh A, Biswal S: Prolonged sulforaphane treatment does not enhance tumorigenesis in oncogenic K-ras and xenograft mouse models of lung cancer. J Carcinog 2012, 11: 8. 10.4103/1477-3163.98459PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Biswas S, Hwang JW, Kirkham PA, Rahman I: Pharmacologial and dietary antioxidant therapies for chronic obstructive pulmonary disease. Curr Med Chem 2013, 20: 1496–1530. 10.2174/0929867311320120004PubMedCrossRef Biswas S, Hwang JW, Kirkham PA, Rahman I: Pharmacologial and dietary antioxidant therapies for chronic obstructive pulmonary disease. Curr Med Chem 2013, 20: 1496–1530. 10.2174/0929867311320120004PubMedCrossRef
13.
Zurück zum Zitat Clarke JD, Hsu A, Williams DE, Dashwood RH, Stevens JF, Yamamoto M, Ho E: Metabolism and tissue distribution of sulforaphane in Nrf2 knockout and wild-type mice. Pharm Res 2011, 28: 3171–3179. 10.1007/s11095-011-0500-zPubMedCentralPubMedCrossRef Clarke JD, Hsu A, Williams DE, Dashwood RH, Stevens JF, Yamamoto M, Ho E: Metabolism and tissue distribution of sulforaphane in Nrf2 knockout and wild-type mice. Pharm Res 2011, 28: 3171–3179. 10.1007/s11095-011-0500-zPubMedCentralPubMedCrossRef
14.
Zurück zum Zitat Weissenberger J, Priester M, Bernreuther C, Rakel S, Glatzel M, Seifert V, Kögel D: Dietary curcumin attenuates glioma growth in a syngeneic mouse model by inhibition of the JAK1,2/STAT3 signaling pathway. Clin Cancer Res 2010, 16: 5781–5795. 10.1158/1078-0432.CCR-10-0446PubMedCrossRef Weissenberger J, Priester M, Bernreuther C, Rakel S, Glatzel M, Seifert V, Kögel D: Dietary curcumin attenuates glioma growth in a syngeneic mouse model by inhibition of the JAK1,2/STAT3 signaling pathway. Clin Cancer Res 2010, 16: 5781–5795. 10.1158/1078-0432.CCR-10-0446PubMedCrossRef
15.
Zurück zum Zitat Xue Y, Ren H, Xiao W, Chu Z, Lee JJ, Mao L: Antitumor activity of AZ64 via G2/M arrest in non-small cell lung cancer. Int J Oncol 2012, 41: 1798–1808.PubMed Xue Y, Ren H, Xiao W, Chu Z, Lee JJ, Mao L: Antitumor activity of AZ64 via G2/M arrest in non-small cell lung cancer. Int J Oncol 2012, 41: 1798–1808.PubMed
16.
Zurück zum Zitat Tang L, Gao Y, Yan F, Tang J: Evaluation of cyclin-dependent kinase-like 1 expression in breast cancer tissues and its regulation in cancer cell growth. Cancer Biother Radiopharm 2012, 27: 392–398. 10.1089/cbr.2012.1198PubMedCrossRef Tang L, Gao Y, Yan F, Tang J: Evaluation of cyclin-dependent kinase-like 1 expression in breast cancer tissues and its regulation in cancer cell growth. Cancer Biother Radiopharm 2012, 27: 392–398. 10.1089/cbr.2012.1198PubMedCrossRef
17.
Zurück zum Zitat Zhang X, Jia S, Yang S, Yang Y, Yang T, Yang Y: Arsenic trioxide induces G2/M arrest in hepatocellular carcinoma cells by increasing the tumor suppressor PTEN expression. J Cell Biochem 2012, 113: 3528–3535. 10.1002/jcb.24230PubMedCrossRef Zhang X, Jia S, Yang S, Yang Y, Yang T, Yang Y: Arsenic trioxide induces G2/M arrest in hepatocellular carcinoma cells by increasing the tumor suppressor PTEN expression. J Cell Biochem 2012, 113: 3528–3535. 10.1002/jcb.24230PubMedCrossRef
18.
Zurück zum Zitat Oliveras-Ferraros C, Fernández-Arroyo S, Vazquez-Martin A, Lozano-Sánchez J, Cufí S, Joven J, Micol V, Fernández-Gutiérrez A, Segura-Carretero A, Menendez JA: Crude phenolic extracts from extra virgin olive oil circumvent de novo breast cancer resistance to HER1/HER2-targeting drugs by inducing GADD45-sensed cellular stress, G2/M arrest and hyperacetylation of Histone H3. Int J Oncol 2011, 38: 1533–1547.PubMed Oliveras-Ferraros C, Fernández-Arroyo S, Vazquez-Martin A, Lozano-Sánchez J, Cufí S, Joven J, Micol V, Fernández-Gutiérrez A, Segura-Carretero A, Menendez JA: Crude phenolic extracts from extra virgin olive oil circumvent de novo breast cancer resistance to HER1/HER2-targeting drugs by inducing GADD45-sensed cellular stress, G2/M arrest and hyperacetylation of Histone H3. Int J Oncol 2011, 38: 1533–1547.PubMed
19.
Zurück zum Zitat Shih RS, Wong SH, Schoene NW, Zhang JJ, Lei KY: Enhanced Gadd45 expression and delayed G2/M progression are p53-dependent in zinc-supplemented human bronchial epithelial cells. Exp Biol Med 2010, 235: 932–940. 10.1258/ebm.2010.010076CrossRef Shih RS, Wong SH, Schoene NW, Zhang JJ, Lei KY: Enhanced Gadd45 expression and delayed G2/M progression are p53-dependent in zinc-supplemented human bronchial epithelial cells. Exp Biol Med 2010, 235: 932–940. 10.1258/ebm.2010.010076CrossRef
20.
Zurück zum Zitat Drews-Elger K, Ortells MC, Rao A, López-Rodriguez C, Aramburu J: The transcription factor NFAT5 is required for cyclin expression and cell cycle progression in cells exposed to hypertonic stress. PLoS One 2009, 4: e5245. 10.1371/journal.pone.0005245PubMedCentralPubMedCrossRef Drews-Elger K, Ortells MC, Rao A, López-Rodriguez C, Aramburu J: The transcription factor NFAT5 is required for cyclin expression and cell cycle progression in cells exposed to hypertonic stress. PLoS One 2009, 4: e5245. 10.1371/journal.pone.0005245PubMedCentralPubMedCrossRef
21.
Zurück zum Zitat Davis R, Singh KP, Kurzrock R, Shankar S: Sulforaphane inhibits angiogenesis through activation of FOXO transcription factors. Oncol Rep 2009, 22: 1473–1478.PubMed Davis R, Singh KP, Kurzrock R, Shankar S: Sulforaphane inhibits angiogenesis through activation of FOXO transcription factors. Oncol Rep 2009, 22: 1473–1478.PubMed
Metadaten
Titel
Sulforaphane induced cell cycle arrest in the G2/M phase via the blockade of cyclin B1/CDC2 in human ovarian cancer cells
verfasst von
Chi-Chang Chang
Chao-Ming Hung
Yun-Ru Yang
Mon-Juan Lee
Yi-Chiang Hsu
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Journal of Ovarian Research / Ausgabe 1/2013
Elektronische ISSN: 1757-2215
DOI
https://doi.org/10.1186/1757-2215-6-41

Weitere Artikel der Ausgabe 1/2013

Journal of Ovarian Research 1/2013 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.