logo logo
Effects of bisphenol A and 17β-estradiol on vascular endothelial growth factor A and its receptor expression in the non-cancer and cancer ovarian cell lines. Ptak Anna,Gregoraszczuk Ewa L Cell biology and toxicology Tumours secrete several pro-angiogenic factors, among which vascular endothelial growth factor (VEGF) and its receptor (VEGF-R) are the most extensively studied but not in ovarian cancer cells. The study was designed to investigate the effect of bisphenol A (BPA) (environmental oestrogen) and of 17β-estradiol (E2) (endogenous estrogen) on the gene (real-time PCR) and protein (Western blotting) expression of VEGF-R2 and VEGF-A in human non-cancer (HOSEpiC) and ovarian cancer cell lines (SKOV-3 and OVCAR-3). In addition, VEGF-A levels were measured in culture supernatants using a colorimetric assay. Cells were exposed to BPA (1, 40 and 100 nM) or 17β-estradiol (0.1, 10 and 40 nM) for 3 to 48 h. Since differential expression levels of basal oestrogen receptor (ERα and ERβ) between non-cancer and cancer cell lines may affect the response to oestrogens, receptor expression was measured both at the gene and protein levels. Basal ERβ expression was similar in all cell lines, and ERα expression was significantly higher in the SKOV-3 cell line. Basal VEGF-R2 expression was higher in cancer than non-cancer cell lines, and in contrast, VEGF-A expression was significantly lower in both SKOV-3 and OVCAR-3 cancer cell lines. Exposure of non-cancer cells to BPA and E2 was associated with a significant increase in VEGF-R2 expression but had no effect on VEGF-A expression or secretion. In contrast, exposure of cancer cells to BPA, but not E2, increased VEGF-R2 and VEGF-A expression and secretion. In conclusion, (1) BPA and E2 regulated VEGF-R2 and VEGF-A expression differently in non-cancer and cancer cells, and (2) BPA has a direct stimulatory effect on VEGF-R2 and VEGF-A expression in both, while E2 appears to be uninvolved in the regulation of VEGF-R2 and VEGF-A expression in cancer cells. Graphical Abstract A schematic representation showing BPA and E2 action on VEGF-R2 and VEGF-A expression in non-cancer (HOSEpiC) and cancer cells (SKOV-3, OVCAR-3). 10.1007/s10565-015-9303-z
Circulating estrogens and postmenopausal ovarian and endometrial cancer risk among current hormone users in the Women's Health Initiative Observational Study. Cancer causes & control : CCC PURPOSE:Menopausal hormone therapy (MHT) use induces alterations in circulating estrogens/estrogen metabolites, which may contribute to the altered risk of reproductive tract cancers among current users. Thus, the current study assessed associations between circulating estrogens/estrogen metabolites and ovarian and endometrial cancer risk among MHT users. METHODS:We conducted a nested case-control study among postmenopausal women using MHT at baseline in the Women's Health Initiative Observational Study (179 ovarian cancers, 396 controls; 230 endometrial cancers, 253 controls). Multivariable logistic regression was utilized to estimate odds ratios and 95% confidence intervals overall and by subtype. RESULTS:Estrogen/estrogen metabolite levels were not associated with overall or serous ovarian cancer risk, examined separately. However, unconjugated estradiol was positively associated with non-serous ovarian cancer risk [quintile 5 vs. quintile 1: 3.01 (1.17-7.73); p-trend = 0.03; p-het < 0.01]. Endometrial cancer risk was unrelated to estrogen/estrogen metabolite levels among women who took combined estrogen/progestin therapy (EPT). CONCLUSIONS:These findings provide novel evidence that may support a heterogeneous hormonal etiology across ovarian cancer subtypes. Circulating estrogens did not influence endometrial cancer risk among women with EPT-induced high-estrogen levels. Larger studies are needed to delineate the relationship between ovarian/endometrial cancer subtypes and estrogen levels in the context of MHT use. 10.1007/s10552-019-01233-8
Identification of Potential Bisphenol A (BPA) Exposure Biomarkers in Ovarian Cancer. Zahra Aeman,Dong Qiduo,Hall Marcia,Jeyaneethi Jeyarooban,Silva Elisabete,Karteris Emmanouil,Sisu Cristina Journal of clinical medicine Endocrine-disrupting chemicals (EDCs) can exert multiple deleterious effects and have been implicated in carcinogenesis. The xenoestrogen Bisphenol A (BPA) that is found in various consumer products has been involved in the dysregulation of numerous signalling pathways. In this paper, we present the analysis of a set of 94 genes that have been shown to be dysregulated in presence of BPA in ovarian cancer cell lines since we hypothesised that these genes might be of biomarker potential. This study sought to identify biomarkers of disease and biomarkers of disease-associated exposure. In silico analyses took place using gene expression data extracted from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases. Differential expression was further validated at protein level using immunohistochemistry on an ovarian cancer tissue microarray. We found that 14 out of 94 genes are solely dysregulated in the presence of BPA, while the remaining 80 genes are already dysregulated (-value < 0.05) in their expression pattern as a consequence of the disease. We also found that seven genes have prognostic power for the overall survival in OC in relation to their expression levels. Out of these seven genes, Keratin 4 (KRT4) appears to be a biomarker of exposure-associated ovarian cancer, whereas Guanylate Binding Protein 5 (GBP5), long intergenic non-protein coding RNA 707 (LINC00707) and Solute Carrier Family 4 Member 11 (SLC4A11) are biomarkers of disease. BPA can exert a plethora of effects that can be tissue- or cancer-specific. Our in silico findings generate a hypothesis around biomarkers of disease and exposure that could potentially inform regulation and policy making. 10.3390/jcm10091979
Bisphenol A induce ovarian cancer cell migration via the MAPK and PI3K/Akt signalling pathways. Ptak Anna,Hoffmann Marta,Gruca Izabella,Barć Justyna Toxicology letters Bisphenol A (BPA), is present in a multitude of products, including food and water containers, food can linings, dentistry sealants, and thermal paper. BPA can induce the growth of human ovarian cancer cell lines. Reduction of adhesion and the initiation of metastasis are important events in cancer progression; therefore, this study investigated the effects of BPA (0.1-100nM) on the migration of OVCAR-3 ovarian cancer cells and the expression levels of metalloproteinases (MMPs) and cadherins. The oestrogenic compound 17β-estradiol (40nM) was used as a positive control for estrogenic properties of bisphenol A. BPA stimulated cell migration, and the effect of BPA was similar to that of 17β-estradiol. BPA-induced cell migration was accompanied by up-regulation of the migration-related factors MMP-2, MMP-9 and N-cadherin, but E-cadherin expression and activity was unaffected. The stimulatory effects of BPA on cell migration were abolished by pre-treatment of the cells with inhibitors of the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase pathways (PI3K). In conclusion, the results presented here show that BPA induces OVCAR-3 cells migration by activating MAPK and PI3K/Akt signalling pathways. 10.1016/j.toxlet.2014.07.001
AGK Unleashes CD8 T Cell Glycolysis to Combat Tumor Growth. Zhou Peipei,Chi Hongbo Cell metabolism Elevated glycolytic metabolism is essential for CD8 T cell antitumor function, but cell-intrinsic factors modulating this process remain elusive. In this issue, Hu et al. (2019) show that the lipid kinase acylglycerol kinase (AGK) promotes the glycolytic and functional fitness of CD8 T cells by inactivating PTEN and boosting mTOR activity, thereby promoting antitumor activity. 10.1016/j.cmet.2019.07.008
Bisphenol A and its derivatives decrease expression of chemerin, which reverses its stimulatory action in ovarian cancer cells. Hoffmann Marta,Rak Agnieszka,Ptak Anna Toxicology letters Chemerin is an adipocyte-secreted protein that associates with obesity, inflammation, metabolic dysfunction, and carcinogenesis. Previous studies have shown human granulosa cells to produce bioactive chemerin and its receptor CMKLR1. In the present study, we demonstrated that the mRNA level of chemerin receptor is higher in a granulosa cell tumor cell line than in epithelial cancer cells, whereas chemerin expression and secretion were lower. Various exogenous factors, such as bisphenol A and its halogenated derivatives tetrabromobisphenol A and tetrachlorobisphenol A, can affect adipokine expression. For this reason, we investigated the effects of bisphenol A and its derivatives on the expression of chemerin and its receptor. At low nanomolar concentrations, BPA, TBBPA, and TCBPA decreased chemerin expression and secretion only in granulosa cell tumor COV434 cells by both peroxisome proliferator-activated receptor γ and estrogen receptor signaling pathways. Chemerin treatment had no effect on proliferation of ovarian non-cancer and cancer cell lines. However, we also found evidence to support the inhibition of BPA- and TBBPA-induced cell proliferation by chemerin. Taken together, our results indicate for the first time that BPA and its derivatives down-regulate chemerin expression, which can suppress the ability of BPA to induce proliferation. Moreover, both PPARγ and ERs were involved in the BPA-induced decrease in chemerin expression, and its ratio was crucial to exert these effects. 10.1016/j.toxlet.2018.04.004
Environmental Estrogens and Their Biological Effects through GPER Mediated Signal Pathways. Qie Yu,Qin Weiping,Zhao Keda,Liu Chang,Zhao Lixia,Guo Liang-Hong Environmental pollution (Barking, Essex : 1987) Many environmental chemicals have been found to exert estrogenic effects in cells and experimental animals by activating nuclear receptors such as estrogen receptors and estrogen-related receptors. These compounds include bisphenols, pesticides, polybrominated diphenyl ethers (PBDEs), organophosphate flame retardants, phthalates and metalloestrogens. G protein-coupled estrogen receptor (GPER) exists widely in numerous cells/tissues of human and other vertebrates. A number of studies have demonstrated that GPER plays a vital role in mediating the estrogenic effects of environmental pollutants. Even at very low concentrations, these chemicals may activate GPER pathways, thus affect many aspects of cellular functions including proliferation, metastasis and apoptosis, resulting in cancer progression, cardiovascular disorders, and reproductive dysfunction. This review summarized the environmental occurrence and human exposure levels of these pollutants, and integrated current experimental evidence toward revealing the underlying mechanisms of pollutant-induced cellular dysfunction via GPER. The GPER mediated rapid non-genomic actions play an important role in the process leading to the adverse effects observed in experimental animals and even in human beings. 10.1016/j.envpol.2021.116826
Inflammatory cytokines as key players of apoptosis induced by environmental estrogens in the ovary. Zhang Fa-Li,Kong Li,Zhao Ai-Hong,Ge Wei,Yan Zi-Hui,Li Lan,De Felici Massimo,Shen Wei Environmental research Natural and synthetic environmental estrogens (EEs), interfering with the physiological functions of the body's estrogens, are widespread and are rising much concern for their possible deleterious effects on human and animal health, in particular on reproduction. In fact, increasing evidence indicate that EEs can be responsible for a variety of disfunctions of the reproductive system especially in females such as premature ovarian insufficiency (POI). Because of their great structural diversity, the modes of action of EEs are controversial. One important way through which EEs exert their effects on reproduction is the induction of apoptosis in the ovary. In general, EEs can exert pro-and anti-apoptotic effects by agonizing or antagonizing numerous estrogen-dependent signaling pathways. In the present work, results concerning apoptotic pathways and diseases induced by representative EEs (such as zearalenone, bisphenol A and di-2-ethylhexyl phthalate), in ovaries throughout development are presented into an integrated network. By reviewing and elaborating these studies, we propose inflammatory factors, centered on the production of tumor necrosis factor (TNF), as a major cause of the induction of apoptosis by EEs in the mammalian ovary. As a consequence, potential strategies to prevent such EE effect are suggested. 10.1016/j.envres.2021.111225
Genome-wide association study identifies new susceptibility loci for epithelial ovarian cancer in Han Chinese women. Chen Kexin,Ma Hongxia,Li Lian,Zang Rongyu,Wang Cheng,Song Fengju,Shi Tingyan,Yu Dianke,Yang Ming,Xue Wenqiong,Dai Juncheng,Li Shuang,Zheng Hong,Wu Chen,Zhang Ying,Wu Xiaohua,Li Dake,Xue Fengxia,Li Haixin,Jiang Zhi,Liu Jibin,Liu Yuexin,Li Pei,Tan Wen,Han Jing,Jie Jiang,Hao Quan,Hu Zhibin,Lin Dongxin,Ma Ding,Jia Weihua,Shen Hongbing,Wei Qingyi Nature communications Ovarian cancer is the leading cause of death from gynaecological malignancies worldwide. Here we perform a three-stage genome-wide association study (GWAS) in Han Chinese women to identify risk genetic variants for epithelial ovarian cancer (EOC). We scan 900,015 single-nucleotide polymorphisms (SNPs) in 1,057 EOC cases and 1,191 controls in stage I, and replicate 41 SNPs (P(meta)<10(-4)) in 960 EOC cases and 1,799 controls (stage II), and an additional 492 EOC cases and 1,004 controls (stage III). Finally, we identify two EOC susceptibility loci at 9q22.33 (rs1413299 in COL15A1, P(meta) = 1.88 × 10(-8)) and 10p11.21 (rs1192691 near ANKRD30A, P(meta) = 2.62 × 10(-8)), and two consistently replicated loci at 12q14.2 (rs11175194 in SRGAP1, P(meta) = 1.14 × 10(-7)) and 9q34.2 (rs633862 near ABO and SURF6, P(meta) = 8.57 × 10(-7)) (P<0.05 in all three stages). These results may advance our understanding of genetic susceptibility to EOC. 10.1038/ncomms5682
Discovery of cancer common and specific driver gene sets. Nucleic acids research Cancer is known as a disease mainly caused by gene alterations. Discovery of mutated driver pathways or gene sets is becoming an important step to understand molecular mechanisms of carcinogenesis. However, systematically investigating commonalities and specificities of driver gene sets among multiple cancer types is still a great challenge, but this investigation will undoubtedly benefit deciphering cancers and will be helpful for personalized therapy and precision medicine in cancer treatment. In this study, we propose two optimization models to de novo discover common driver gene sets among multiple cancer types (ComMDP) and specific driver gene sets of one certain or multiple cancer types to other cancers (SpeMDP), respectively. We first apply ComMDP and SpeMDP to simulated data to validate their efficiency. Then, we further apply these methods to 12 cancer types from The Cancer Genome Atlas (TCGA) and obtain several biologically meaningful driver pathways. As examples, we construct a common cancer pathway model for BRCA and OV, infer a complex driver pathway model for BRCA carcinogenesis based on common driver gene sets of BRCA with eight cancer types, and investigate specific driver pathways of the liquid cancer lymphoblastic acute myeloid leukemia (LAML) versus other solid cancer types. In these processes more candidate cancer genes are also found. 10.1093/nar/gkx089
Synergistic effects of combined platelet-activating factor receptor and epidermal growth factor receptor targeting in ovarian cancer cells. Yu Yi,Zhang Mingxing,Zhang Xiaoyan,Cai Qingqing,Hong Shanshan,Jiang Wei,Xu Congjian Journal of hematology & oncology BACKGROUND:Genetic alterations, including the overexpression of epidermal growth factor receptor (EGFR), play a crucial role in ovarian carcinogenesis. To date, EGFR targeting has shown limited antitumor effects in ovarian cancer when administered as monotherapy. We previously identified platelet-activating factor receptor (PAFR) as being overexpressed in ovarian cancer and found that its ligand PAF evoked EGFR phosphorylation. To determine whether PAFR targeting can enhance the antitumor efficacy of EGFR inhibition, we investigated the effects of a PAFR antagonist (WEB2086) in conjunction with an EGFR inhibitor (AG1478). METHODS:The expression of EGFR and PAFR in CAOV-3 and SKOV-3 ovarian cancer cell lines was measured by Western blot and immunocytochemistry. Synergy was determined using isobologram analysis. The effects of combined PAFR and EGFR targeting on both cells were assessed by using CCK-8, transwell, flow cytometry, western blot analysis. In vivo studies were conducted using CAOV-3 cells xenografted in nu/nu mice. RESULTS:Treatment with combination WEB2086 and AG1478 resulted in significantly greater inhibition of proliferation and invasion compared to either drug alone. When examining equipotent combinations of WEB2086 and AG1478 to determine potential synergy, a combination index (CI) of 0.49 was identified for CAOV-3 cells and a CI of 0.58 for SKOV-3 cells indicating synergy. This co-inhibition induced significantly more apoptosis and arrested the cells at G0/G1 phase in both cell lines. The activation of PAFR and/or EGFR induced phosphorylation of the mTOR, AKT, and MAPK pathways. Combined PAFR and EGFR targeting synergistically diminished the expression of PAFR and EGFR phosphorylation and downstream signaling. In vivo studies further verified the antitumor effects of combined PAFR and EGFR targeting in a CAOV-3 xenograft model. CONCLUSIONS:These results suggest that WEB2086 and AG1478 are synergistic in ovarian cancer cells with high expression of both PAFR and EGFR. The presented approach may have important therapeutic implications in the treatment of ovarian cancer patients. 10.1186/1756-8722-7-39
G Protein-Coupled Receptors (GPCRs)-Mediated Calcium Signaling in Ovarian Cancer: Focus on GPCRs activated by Neurotransmitters and Inflammation-Associated Molecules. Predescu Dragoș-Valentin,Crețoiu Sanda Maria,Crețoiu Dragoș,Pavelescu Luciana Alexandra,Suciu Nicolae,Radu Beatrice Mihaela,Voinea Silviu-Cristian International journal of molecular sciences G-coupled protein receptors (GCPR) involve several signaling pathways, some of them being coupled with intracellular calcium (Ca) mobilization. GPCRs were involved in migration, invasion and metastasis of different types of cancers, including ovarian cancer. Many studies have discussed the essential contribution of GPCRs activated by steroid hormones in ovarian cancer. However, ovarian cancer is also associated with altered signals coming from the nervous system, the immune system or the inflammatory environment, in which GPCRs are 'sensing' these molecular signals. Many studies have been oriented so far on ovarian cell lines (most of them being of human cell lines), and only few studies based on animal models or clinical studies have been devoted to the expression changes or functional role of GPCRs in ovarian cancer. In this paper, we review the alterations of GPCRs activated by neurotransmitters (muscarinic receptors, serotonin receptors, dopamine receptors, adrenoceptors) or inflammation-associated molecules (bradykinin receptors, histamine receptors, chemokine receptors) in ovarian cancer and we discuss their potential as histological biomarkers. 10.3390/ijms20225568