1
|
Zhang J, Zhao C, Hu T, Yang J, Wu Q, Wang C, Dong S. The role of PI3K/AKT-related proteins in di(2-ethyl)hexylphthalate-induced BG-1 and MCF-7 cell proliferation, and inhibition by metformin. Immunopharmacol Immunotoxicol 2022; 45:378-385. [DOI: 10.1080/08923973.2022.2151915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Jiatai Zhang
- Department of Environmental Hygiene, Public Health Collage, Harbin Medical University, Harbin, China
| | - Chenyang Zhao
- Department of Environmental Hygiene, Public Health Collage, Harbin Medical University, Harbin, China
| | - Tengteng Hu
- Department of Environmental Hygiene, Public Health Collage, Harbin Medical University, Harbin, China
| | - Jipeng Yang
- Department of Environmental Hygiene, Public Health Collage, Harbin Medical University, Harbin, China
| | - Qian Wu
- Department of Environmental Hygiene, Public Health Collage, Harbin Medical University, Harbin, China
| | - Cheng Wang
- Department of Environmental Hygiene, Public Health Collage, Harbin Medical University, Harbin, China
| | - Shuying Dong
- Department of Environmental Hygiene, Public Health Collage, Harbin Medical University, Harbin, China
| |
Collapse
|
2
|
Pal P, Hales K, Petrik J, Hales DB. Pro-apoptotic and anti-angiogenic actions of 2-methoxyestradiol and docosahexaenoic acid, the biologically derived active compounds from flaxseed diet, in preventing ovarian cancer. J Ovarian Res 2019; 12:49. [PMID: 31128594 PMCID: PMC6535187 DOI: 10.1186/s13048-019-0523-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND We have previously shown that a whole flaxseed supplemented diet decreased the onset and severity of ovarian cancer in the laying hen, the only known animal model of spontaneous ovarian cancer. Flaxseed is rich in omega-3 fatty acids (OM3FA), mostly α-Linoleic acid (ALA), which gets converted to Docosahexaenoic acid (DHA) by the action of delta-6 desaturase enzyme. Ingestion of flaxseed also causes an increase in production of 2-methoxyestradiol (2MeOE2) via the induction of the CYP1A1 pathway of estrogen metabolism. We have previously reported that the flaxseed diet induces apoptosis via p38-MAPK pathway in chicken tumors. The objective of this study was to investigate the effect of the flaxseed diet on ovarian cancer in chickens, focusing on two hallmarks of cancer, apoptosis and angiogenesis. RESULTS The anti-cancer effects of two active biologically derived compounds of flax diet, 2MeOE2 and DHA, were individually tested on human ovarian cancer cells and in vivo by the Chick Chorioallantoic Membrane (CAM) assay. Our results indicate that a flaxseed-supplemented diet promotes apoptosis and inhibits angiogenesis in chicken tumors but not in normal ovaries. 2MeOE2 promotes apoptosis in human ovarian cancer cells, inhibits angiogenesis on CAM and its actions are dependent on the p38-MAPK pathway. DHA does not have any pro-apoptotic effect on human ovarian cancer cells but has strong anti-angiogenic effects as seen on CAM, but not dependent on the p38-MAPK pathway. CONCLUSIONS Dietary flaxseed supplementation promotes a pro-apoptotic and anti-angiogenic effect in ovarian tumors, not in normal ovaries. The biologically derived active compounds from flaxseed diet act through different pathways to elicit their respective anti-cancer effects. A flaxseed-supplemented diet is a promising approach for prevention of ovarian cancer as well as having a significant potential as an adjuvant treatment to supplement chemotherapeutic agents for treatment of advanced stages of ovarian cancer.
Collapse
Affiliation(s)
- Purab Pal
- Department of Physiology, Southern Illinois University, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, IL, 62901, USA
| | - Karen Hales
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Jim Petrik
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Dale Buchanan Hales
- Department of Physiology, Southern Illinois University, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, IL, 62901, USA.
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA.
| |
Collapse
|
3
|
Nunes HC, Scarano WR, Deffune E, Felisbino SL, Porreca I, Delella FK. Bisphenol a and mesenchymal stem cells: Recent insights. Life Sci 2018; 206:22-28. [DOI: 10.1016/j.lfs.2018.05.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 02/08/2023]
|
4
|
Lee GA, Choi KC, Hwang KA. Kaempferol, a phytoestrogen, suppressed triclosan-induced epithelial-mesenchymal transition and metastatic-related behaviors of MCF-7 breast cancer cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 49:48-57. [PMID: 27902959 DOI: 10.1016/j.etap.2016.11.016] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/14/2016] [Accepted: 11/20/2016] [Indexed: 06/06/2023]
Abstract
As a phytoestrogen, kaempferol is known to play a chemopreventive role inhibiting carcinogenesis and cancer progression. In this study, the influences of triclosan, an anti-bacterial agent recently known for an endocrine disrupting chemical (EDC), and kaempferol on breast cancer progression were examined by measuring their effects on epithelial-mesenchymal transition (EMT) and metastatic-related behaviors of MCF-7 breast cancer cells. Morphological changes of MCF-7 cells were observed, and a wound-healing assay was performed after the treatment of triclosan and kaempferol. The effects of triclosan and kaempferol on protein expression of EMT-related markers such as E-cadherin, N-cadherin, Snail, and Slug and metastasis-related markers such as cathepsin B, D, MMP-2 and -9 were investigated by Western blot assay. In microscopic observations, triclosan (10-6M) or E2 (10-9M) induced transition to mesenchymal phenotype of MCF-7 cells compared with the control. Co-treatment of ICI 182,780 (10-8M), an ER antagonist, or kaempferol (25μM) with E2 or triclosan restored the cellular morphology to an epithelial phenotype. In a wound-healing scratch and a transwell migration assay, triclosan enhanced migration and invasion of MCF-7 cells, but co-treatment of kaempferol or ICI 182,780 reduced the migration and invasion ability of MCF-7 cells to the control level. In addition, kaempferol effectively suppressed E2 or triclosan-induced protein expressions of EMT and metastasis promoting markers. Taken together, triclosan may be a distinct xenoestrogenic EDC to promote EMT, migration, and invasion of MCF-7 breast cancer cells through ER. On the other hand, kaempferol can be an alternative chemopreventive agent to effectively suppress the metastatic behavior of breast cancer induced by an endogenous estrogen as well as exogenous xenoestrogenic compounds including triclosan.
Collapse
Affiliation(s)
- Geum-A Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| |
Collapse
|
5
|
Tamir A, Jag U, Sarojini S, Schindewolf C, Tanaka T, Gharbaran R, Patel H, Sood A, Hu W, Patwa R, Blake P, Chirina P, Oh Jeong J, Lim H, Goy A, Pecora A, Suh KS. Kallikrein family proteases KLK6 and KLK7 are potential early detection and diagnostic biomarkers for serous and papillary serous ovarian cancer subtypes. J Ovarian Res 2014; 7:109. [PMID: 25477184 PMCID: PMC4271347 DOI: 10.1186/s13048-014-0109-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 11/11/2014] [Indexed: 12/13/2022] Open
Abstract
Background Early detection of ovarian cancer remains a challenge due to widespread metastases and a lack of biomarkers for early-stage disease. This study was conducted to identify relevant biomarkers for both laparoscopic and serum diagnostics in ovarian cancer. Methods Bioinformatics analysis and expression screening in ovarian cancer cell lines were employed. Selected biomarkers were further validated in bio-specimens of diverse cancer types and ovarian cancer subtypes. For non-invasive detection, biomarker proteins were evaluated in serum samples from ovarian cancer patients. Results Two kallikrein (KLK) serine protease family members (KLK6 and KLK7) were found to be significantly overexpressed relative to normal controls in most of the ovarian cancer cell lines examined. Overexpression of KLK6 and KLK7 mRNA was specific to ovarian cancer, in particular to serous and papillary serous subtypes. In situ hybridization and histopathology further confirmed significantly elevated levels of KLK6 and KLK7 mRNA and proteins in tissue epithelium and a lack of expression in neighboring stroma. Lastly, KLK6 and KLK7 protein levels were significantly elevated in serum samples from serous and papillary serous subtypes in the early stages of ovarian cancer, and therefore could potentially decrease the high “false negative” rates found in the same patients with the common ovarian cancer biomarkers human epididymis protein 4 (HE4) and cancer antigen 125 (CA-125). Conclusion KLK6 and KLK7 mRNA and protein overexpression is directly associated with early-stage ovarian tumors and can be measured in patient tissue and serum samples. Assays based on KLK6 and KLK7 expression may provide specific and sensitive information for early detection of ovarian cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13048-014-0109-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ayala Tamir
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Ushma Jag
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Sreeja Sarojini
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Craig Schindewolf
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Takemi Tanaka
- Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Rajendra Gharbaran
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Hiren Patel
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Anil Sood
- Departments of Gynecologic Oncology and Cancer Biology, MD Anderson Cancer Center, University of Texas, Houston, TX, 77030, USA.
| | - Wei Hu
- Departments of Gynecologic Oncology and Cancer Biology, MD Anderson Cancer Center, University of Texas, Houston, TX, 77030, USA.
| | - Ruzeen Patwa
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Patrick Blake
- Sophic Systems Alliance, Inc, Rockville, MD, 20850, USA.
| | - Polina Chirina
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Jin Oh Jeong
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Heejin Lim
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Andre Goy
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - Andrew Pecora
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| | - K Stephen Suh
- The Genomics and Biomarkers Program, The John Theurer Cancer Center, Hackensack University Medical Center, D. Jurist Research Building, 40 Prospect Avenue, Hackensack, NJ, 07601, USA.
| |
Collapse
|
6
|
Kasiappan R, Sun Y, Lungchukiet P, Quarni W, Zhang X, Bai W. Vitamin D suppresses leptin stimulation of cancer growth through microRNA. Cancer Res 2014; 74:6194-204. [PMID: 25252917 DOI: 10.1158/0008-5472.can-14-1702] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity is a pandemic and major risk factor for cancers. The reduction of obesity would have been an effective strategy for cancer prevention, but the reality is that worldwide obesity has kept increasing for decades, remaining a major avoidable cancer risk secondary only to smoke. The present studies suggest that vitamin D may be an effective agent to reduce obesity-associated cancer risks in women. Molecular analyses showed that leptin increased human telomerase reverse transcriptase (hTERT) mRNA expression and cell growth through estrogen receptor-α (ERα) activation in ovarian cancer cells, which was suppressed by 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3]. The suppression was compromised when miR-498 induction by the hormone was depleted with microRNA (miRNA) sponges. In mice, high-fat diet (HFD) stimulation of ovarian tumor growth was remarkably suppressed by 1,25(OH)2D3 analogue EB1089, which was also compromised by miR-498 sponges. EB1089 did not alter HFD-induced increase in serum leptin levels but increased miR-498 and decreased the diet-induced hTERT expression in tumors. Quantitative RT-PCR analyses revealed an inverse correlation between hTERT mRNA and miR-498 in response to 1,25(OH)2D3 in estrogen-sensitive ovarian, endometrial, and breast cancers. The studies suggest that miR-498-mediated hTERT downregulation is a key event mediating the anti-leptin activity of 1,25(OH)2D3 in estrogen-sensitive tumors in women.
Collapse
Affiliation(s)
- Ravi Kasiappan
- Department of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, Florida
| | - Yuefeng Sun
- Department of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, Florida
| | - Panida Lungchukiet
- Department of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, Florida
| | - Waise Quarni
- Department of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, Florida
| | - Xiaohong Zhang
- Department of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, Florida. Department of Oncological Sciences, University of South Florida College of Medicine, Tampa, Florida. Program of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Wenlong Bai
- Department of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, Florida. Department of Oncological Sciences, University of South Florida College of Medicine, Tampa, Florida. Program of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center, Tampa, Florida.
| |
Collapse
|
7
|
Wang S, Aarts JMMJG, de Haan LHJ, Argyriou D, Peijnenburg AACM, Rietjens IMCM, Bovee TFH. Towards an integratedin vitrostrategy for estrogenicity testing. J Appl Toxicol 2013; 34:1031-40. [DOI: 10.1002/jat.2928] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/11/2013] [Accepted: 08/04/2013] [Indexed: 12/30/2022]
Affiliation(s)
- Si Wang
- Division of Toxicology; Wageningen University and Research Centre; Tuinlaan 5 6703 HE Wageningen The Netherlands
- Business Unit of Toxicology & Bioassays, RIKILT - Institute of Food Safety; Wageningen University and Research Centre; Akkermaalsbos 2 6708 WB Wageningen The Netherlands
| | - Jac M. M. J. G. Aarts
- Business Unit of Toxicology & Bioassays, RIKILT - Institute of Food Safety; Wageningen University and Research Centre; Akkermaalsbos 2 6708 WB Wageningen The Netherlands
| | - Laura H. J. de Haan
- Division of Toxicology; Wageningen University and Research Centre; Tuinlaan 5 6703 HE Wageningen The Netherlands
| | - Dimitrios Argyriou
- Division of Toxicology; Wageningen University and Research Centre; Tuinlaan 5 6703 HE Wageningen The Netherlands
| | - Ad A. C. M. Peijnenburg
- Business Unit of Toxicology & Bioassays, RIKILT - Institute of Food Safety; Wageningen University and Research Centre; Akkermaalsbos 2 6708 WB Wageningen The Netherlands
| | - Ivonne M. C. M. Rietjens
- Division of Toxicology; Wageningen University and Research Centre; Tuinlaan 5 6703 HE Wageningen The Netherlands
| | - Toine F. H. Bovee
- Business Unit of Toxicology & Bioassays, RIKILT - Institute of Food Safety; Wageningen University and Research Centre; Akkermaalsbos 2 6708 WB Wageningen The Netherlands
| |
Collapse
|
8
|
Guo R, Abdelmohsen K, Morin PJ, Gorospe M. Novel MicroRNA Reporter Uncovers Repression of Let-7 by GSK-3β. PLoS One 2013; 8:e66330. [PMID: 23840442 PMCID: PMC3694080 DOI: 10.1371/journal.pone.0066330] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 05/03/2013] [Indexed: 02/04/2023] Open
Abstract
Several members of the let-7 microRNA family are downregulated in ovarian and other cancers. They are thought to act as tumor suppressors by lowering growth-promoting and anti-apoptotic proteins. In order to measure cellular let-7 levels systematically, we have developed a highly sensitive let-7 reporter assay system based on the expression of a chimeric mRNA that contains the luciferase coding region and a 3′-untranslated region (UTR) bearing two let-7-binding sites. In cells expressing the reporter construct, termed pmirGLO-let7, luciferase activity was high when let-7 was absent, while luciferase activity was low when let-7 levels were elevated. The ovarian cancer cell lines BG-1 and UCI-101 were transfected with the let-7 reporter and surveyed with a library of kinase inhibitors in order to identify pathways affecting let-7 activity. Among the inhibitors causing changes in endogenous let-7 abundance, the lowering of glycogen synthase kinase 3 (GSK-3)β function specifically increased let-7 levels and lowered luciferase activity. Similarly, silencing GSK-3β increased both mature and primary-let-7 levels in BG-1 cells, and decreased BG-1 cell survival. Further studies identified p53 as a downstream effector of the GSK-3β-mediated repression of let-7 biosynthesis. Our studies highlight GSK-3β as a novel therapeutic target in ovarian tumorigenesis.
Collapse
Affiliation(s)
- Rong Guo
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, Maryland, United States of America
| | - Kotb Abdelmohsen
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, Maryland, United States of America
| | - Patrice J. Morin
- Laboratory of Molecular Biology and Immunology, National Institute on Aging-Intramural Research Program, NIH, Baltimore, Maryland, United States of America
- American Association for Cancer Research, Philadelphia, Pennsylvania, United States of America
- * E-mail: (MG); (PM)
| | - Myriam Gorospe
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, Maryland, United States of America
- * E-mail: (MG); (PM)
| |
Collapse
|
9
|
Hwang KA, Kang NH, Yi BR, Lee HR, Park MA, Choi KC. Genistein, a soy phytoestrogen, prevents the growth of BG-1 ovarian cancer cells induced by 17β-estradiol or bisphenol A via the inhibition of cell cycle progression. Int J Oncol 2013; 42:733-40. [PMID: 23229410 DOI: 10.3892/ijo.2012.1719] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 11/02/2012] [Indexed: 11/06/2022] Open
Abstract
An endocrine disrupting chemical (EDC) is a global health concern. In this study, we examined the effects of genistein (GEN) on bisphenol A (BPA) or 17β-estradiol (E2)-induced cell growth and gene alterations of BG-1 ovarian cancer cells expressing estrogen receptors (ERs). In an in vitro cell viability assay, E2 or BPA significantly increased the growth of BG-1 cells. This increased proliferative activity was reversed by treatment with ICI 182,780, a well-known ER antagonist, while cell proliferation was further promoted in the presence of propyl pyrazole triol (PPT), an ERα agonist. These results imply that cell proliferation increased by E2 or BPA was mediated by ERs, particularly ERα. BPA clearly acted as a xenoestrogen in BG-1 ovarian cancer cells by mimicking E2 action. In contrast, GEN effectively suppressed BG-1 cell proliferation promoted by E2 or BPA by inhibiting cell cycle progression. E2 and BPA increased the expression of cyclin D1, a factor responsible for the G1/S cell cycle transition. They also decreased the expression of p21, a potent cyclin-dependent kinase (CDK) inhibitor that arrests the cell cycle in G1 phase, and promoted the proliferation of BG-1 cells. As shown by its repressive effect on cell growth, GEN decreased the expression of cyclin D1 augmented by E2 or BPA. On the other hand, GEN increased the p21 expression downregulated by E2 or BPA. Collectively, our findings suggest that GEN, a dietary phytoestrogen, has an inhibitory effect on the growth of estrogen-dependent cancers promoted by E2 or BPA.
Collapse
Affiliation(s)
- Kyung-A Hwang
- Laboratory of Veterinary Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, Republic of Korea
| | | | | | | | | | | |
Collapse
|
10
|
Bossard C, Busson M, Vindrieux D, Gaudin F, Machelon V, Brigitte M, Jacquard C, Pillon A, Balaguer P, Balabanian K, Lazennec G. Potential role of estrogen receptor beta as a tumor suppressor of epithelial ovarian cancer. PLoS One 2012; 7:e44787. [PMID: 22970307 PMCID: PMC3435304 DOI: 10.1371/journal.pone.0044787] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 08/13/2012] [Indexed: 11/28/2022] Open
Abstract
Ovarian cancer is the gynecological cancer exhibiting the highest morbidity and improvement of treatments is still required. Previous studies have shown that Estrogen-receptor beta (ERβ) levels decreased along with ovarian carcinogenesis. Here, we present evidence that reintroduction of ERβ in BG-1 epithelial ovarian cancer cells, which express ERα, leads in vitro to a decrease of basal and estradiol-promoted cell proliferation. ERβ reduced the frequency of cells in S phase and increased the one of cells in G2/M phase. At the molecular level, we found that ERβ downregulated total retinoblastoma (Rb), phosphorylated Rb and phospho-AKT cellular content as well as cyclins D1 and A2. In addition, ERβ had a direct effect on ERα, by strongly inhibiting its expression and activity, which could explain part of the anti-proliferative action of ERβ. By developing a novel preclinical model of ovarian cancer based on a luminescent orthotopic xenograft in athymic Nude mice, we further revealed that ERβ expression reduces tumor growth and the presence of tumor cells in sites of metastasis, hence resulting in improved survival of mice. Altogether, these findings unveil a potential tumor-suppressor role of ERβ in ovarian carcinogenesis, which could be of potential clinical relevance for the selection of the most appropriate treatment for patients.
Collapse
Affiliation(s)
- Carine Bossard
- Institut National de la Santé et de la Recherche Médicale (INSERM), U844, University of Montpellier I, Montpellier, F-34091, France
| | - Muriel Busson
- Institut National de la Santé et de la Recherche Médicale (INSERM), U844, University of Montpellier I, Montpellier, F-34091, France
| | - David Vindrieux
- Institut National de la Santé et de la Recherche Médicale (INSERM), U844, University of Montpellier I, Montpellier, F-34091, France
| | - Françoise Gaudin
- Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LERMIT), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR_S996, Univ. Paris-Sud, Clamart, France
| | - Véronique Machelon
- Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LERMIT), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR_S996, Univ. Paris-Sud, Clamart, France
| | - Madly Brigitte
- Institut National de la Santé et de la Recherche Médicale (INSERM), U844, University of Montpellier I, Montpellier, F-34091, France
| | - Carine Jacquard
- Institut National de la Santé et de la Recherche Médicale (INSERM), U844, University of Montpellier I, Montpellier, F-34091, France
| | - Arnaud Pillon
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Montpellier, France
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Montpellier, France
| | - Karl Balabanian
- Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LERMIT), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR_S996, Univ. Paris-Sud, Clamart, France
| | - Gwendal Lazennec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U844, University of Montpellier I, Montpellier, F-34091, France
- * E-mail:
| |
Collapse
|
11
|
Wang S, Aarts JMMJG, Evers NM, Peijnenburg AACM, Rietjens IMCM, Bovee TFH. Proliferation assays for estrogenicity testing with high predictive value for the in vivo uterotrophic effect. J Steroid Biochem Mol Biol 2012; 128:98-106. [PMID: 22138011 DOI: 10.1016/j.jsbmb.2011.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 11/15/2011] [Accepted: 11/16/2011] [Indexed: 12/22/2022]
Abstract
Proliferation assays based on human cell lines are the most used in vitro tests to determine estrogenic properties of compounds. Our objective was to characterise to what extent these in vitro tests provide alternatives for the in vivo Allen and Doisy test, a uterotrophic assay in immature or ovariectomised rodents with uterus weight as a crucial read-out parameter. In the present study four different human cell lines derived from three different female estrogen-sensitive tissues, i.e. breast (MCF-7/BOS and T47D), endometrial (ECC-1) and ovarian (BG-1) cells, were characterised by investigating their relative ERα and ERβ amounts, as the ERα/ERβ ratio is a dominant factor determining their estrogen-dependent proliferative responses. All four cell lines clearly expressed the ERα type and a very low but detectable amount of ERβ on both the mRNA and protein level, with the T47D cell line expressing the highest level of the ERβ type. Subsequently, a set of reference compounds representing different modes of estrogen action and estrogenic potency were used to investigate the proliferative response in the four cell lines, to determine which cell line most accurately predicts the effect observed in vivo. All four cell lines revealed a reasonable to good correlation with the in vivo uterotrophic effect, with the correlation being highest for the MCF-7/BOS cell line (R²=0.85). The main differences between the in vivo uterotrophic assay and the in vitro proliferation assays were observed for tamoxifen and testosterone. The proliferative response of the MCF-7/BOS cells to testosterone was partially caused by its conversion to estradiol by aromatase or via androstenedione to estrone. It is concluded that of the four cell lines tested, the best assay to include in an integrated testing strategy for replacement of the in vivo uterotrophic assay is the human MCF-7/BOS breast cancer cell line.
Collapse
Affiliation(s)
- Si Wang
- Division of Toxicology, Wageningen University and Research Centre, Tuinlaan 5, 6703 HE Wageningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
12
|
Schröpfer A, Kammerer U, Kapp M, Dietl J, Feix S, Anacker J. Expression pattern of matrix metalloproteinases in human gynecological cancer cell lines. BMC Cancer 2010; 10:553. [PMID: 20942921 PMCID: PMC2964638 DOI: 10.1186/1471-2407-10-553] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 10/13/2010] [Indexed: 02/22/2023] Open
Abstract
Background Matrix metalloproteinases (MMPs) are involved in the degradation of protein components of the extracellular matrix and thus play an important role in tumor invasion and metastasis. Their expression is related to the progression of gynecological cancers (e.g. endometrial, cervical or ovarian carcinoma). In this study we investigated the expression pattern of the 23 MMPs, currently known in humans, in different gynecological cancer cell lines. Methods In total, cell lines from three endometrium carcinomas (Ishikawa, HEC-1-A, AN3 CA), three cervical carcinomas (HeLa, Caski, SiHa), three chorioncarcinomas (JEG, JAR, BeWo), two ovarian cancers (BG-1, OAW-42) and one teratocarcinoma (PA-1) were examined. The expression of MMPs was analyzed by RT-PCR, Western blot and gelatin zymography. Results We demonstrated that the cell lines examined can constitutively express a wide variety of MMPs on mRNA and protein level. While MMP-2, -11, -14 and -24 were widely expressed, no expression was seen for MMP-12, -16, -20, -25, -26, -27 in any of the cell lines. A broad range of 16 MMPs could be found in the PA1 cells and thus this cell line could be used as a positive control for general MMP experiments. While the three cervical cancer cell lines expressed 10-14 different MMPs, the median expression in endometrial and choriocarcinoma cells was 7 different enzymes. The two investigated ovarian cancer cell lines showed a distinctive difference in the number of expressed MMPs (2 vs. 10). Conclusions Ishikawa, Caski, OAW-42 and BeWo cell lines could be the best choice for all future experiments on MMP regulation and their role in endometrial, cervical, ovarian or choriocarcinoma development, whereas the teratocarcinoma cell line PA1 could be used as a positive control for general MMP experiments.
Collapse
Affiliation(s)
- Andrea Schröpfer
- Department of Obstetrics and Gynecology, University of Wuerzburg, Josef-Schneider Str 4, 97080 Wuerzburg, Germany
| | | | | | | | | | | |
Collapse
|
13
|
Bendrik C, Karlsson L, Dabrosin C. Increased endostatin generation and decreased angiogenesis via MMP-9 by tamoxifen in hormone dependent ovarian cancer. Cancer Lett 2009; 292:32-40. [PMID: 19944523 DOI: 10.1016/j.canlet.2009.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 10/30/2009] [Accepted: 11/03/2009] [Indexed: 11/17/2022]
Abstract
There are several similarities between breast and ovarian cancer but anti-estrogen treatment is rarely used in ovarian cancer. We have previously shown that the most widely used anti-estrogen tamoxifen increased MMP-9 activity and endostatin generation in breast cancer. Here, we show that tamoxifen exposure of highly hormone responsive ovarian cancer cells decreased proliferation, and increased MMP-9 activity leading to increased levels of endostatin both in cell culture in vitro and in solid tumors of nude mice. Tamoxifen exposed tumors also exhibited significantly decreased tumor growth and vascularisation. Moreover, in ascites from ovarian cancer patients, MMP-9 was undetectable in majority of cases but a significant correlation of MMP-2 and endostatin was found. The effects on MMPs and endostatin generation are previously unknown mechanisms of estradiol and tamoxifen in ovarian cancer, which may have therapeutic implications in future anti-cancer options of hormone dependent ovarian cancer.
Collapse
Affiliation(s)
- Christina Bendrik
- Faculty of Health Sciences, Department of Clinical and Experimental Medicine, Division of Oncology, Linköping University, Linköping, Sweden
| | | | | |
Collapse
|
14
|
Vicus D, Rosen B, Lubinski J, Domchek S, Kauff ND, Lynch HT, Isaacs C, Tung N, Sun P, Narod SA. Tamoxifen and the risk of ovarian cancer in BRCA1 mutation carriers. Gynecol Oncol 2009; 115:135-137. [PMID: 19577280 DOI: 10.1016/j.ygyno.2009.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 06/09/2009] [Accepted: 06/10/2009] [Indexed: 11/26/2022]
Abstract
OBJECTIVE BRCA1 mutation carriers have a high rate of both breast and ovarian cancer. Tamoxifen is a selective estrogen receptor modulator (SERM), which is used for the treatment of primary breast cancer and for the prevention of contralateral breast cancer. Our objective is to assess if tamoxifen treatment is associated with an increase in the subsequent risk of ovarian cancer among women with a BRCA1 mutation. METHODS A matched case-control study was performed. Cases were 154 women with ovarian cancer and a previous history of breast cancer. Controls were 560 women with no ovarian cancer and a history of breast cancer. All cases and controls carry a deleterious BRCA1 mutation. Cases and controls were matched for year of birth, age at diagnosis of breast cancer and country of residence. The effect of tamoxifen treatment on the risk of subsequent ovarian cancer was estimated using conditional logistic regression. RESULTS The unadjusted odds ratio for ovarian cancer, given previous tamoxifen treatment was 0.89 (95% CI 0.54-1.49, p=0.66). After adjusting for other treatments, the odds ratio was 0.78 (95% CI 0.46-1.33, p=0.36). CONCLUSION Tamoxifen treatment for breast cancer does not appear to increase the risk of ovarian cancer in BRCA1 mutation carriers.
Collapse
Affiliation(s)
- Danielle Vicus
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Toronto, Ontario, Canada; Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
| | - Barry Rosen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Toronto, Ontario, Canada
| | - Jan Lubinski
- Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Susan Domchek
- Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Noah D Kauff
- Clinical Genetics and Gynecology Services, Memorial Sloan-Kettering Cancer Centre, New York, NY, USA
| | - Henry T Lynch
- Department of Preventive Medicine and Public Health, Creighton University School of Medicine, Omaha, NE, USA
| | - Claudine Isaacs
- Lombardi Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Nadine Tung
- Beth Israel Deaconess Hospital, Boston, MA, USA
| | - Ping Sun
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada.
| | | |
Collapse
|
15
|
Eckstein N, Servan K, Hildebrandt B, Pölitz A, von Jonquières G, Wolf-Kümmeth S, Napierski I, Hamacher A, Kassack MU, Budczies J, Beier M, Dietel M, Royer-Pokora B, Denkert C, Royer HD. Hyperactivation of the insulin-like growth factor receptor I signaling pathway is an essential event for cisplatin resistance of ovarian cancer cells. Cancer Res 2009; 69:2996-3003. [PMID: 19318572 DOI: 10.1158/0008-5472.can-08-3153] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Platinum plays a central role in the therapy of ovarian cancer, and the emergence of platinum resistance is a major obstacle for clinical management of the disease. We treated A2780 ovarian cancer cells by weekly cycles of cisplatin over a period of 6 months and unveiled that enhanced insulin-like growth factor I receptor (IGF-IR) expression and autocrine IGF-I are associated with hyperactivation of the IGF-IR and phosphatidylinositol-3-OH kinase (PI3K) pathways in cisplatin-resistant cells. IGF-IR expression levels increased during treatment cycles and correlated with cisplatin resistance. Purified IGF-I induced cisplatin resistance in diverse ovarian cancer cell lines, and small molecule inhibitors proved that IGF-IR and PI3K are essential for cisplatin resistance. Similar results were obtained with BG-1 ovarian cancer cells. Cytogenetic and array comparative genomic hybridization analyses revealed selection and de novo formation of chromosomal alterations during resistance development. An analysis of gene expression profiles of primary ovarian carcinomas identified the regulatory subunit PIK3R2 of PI3-kinase as a significant negative prognosis factor for ovarian cancer. We conclude that targeting the IGF-IR and the PI3K pathways is a promising new strategy to treat cisplatin-resistant ovarian carcinomas.
Collapse
Affiliation(s)
- Niels Eckstein
- Center of Advanced European Studies and Research (caesar), Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Hurt EM, Saykally JN, Anose BM, Kalli KR, Sanders MM. Expression of the ZEB1 (deltaEF1) transcription factor in human: additional insights. Mol Cell Biochem 2008; 318:89-99. [PMID: 18622689 DOI: 10.1007/s11010-008-9860-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Accepted: 06/25/2008] [Indexed: 01/13/2023]
Abstract
The zinc finger E-box binding transcription factor ZEB1 (deltaEF1/Nil-2-a/AREB6/zfhx1a/TCF8/zfhep/BZP) is emerging as an important regulator of the epithelial to mesenchymal transitions (EMT) required for development and cancer metastasis. ZEB1 promotes EMT by repressing genes contributing to the epithelial phenotype while activating those associated with the mesenchymal phenotype. TCF8 (zfhx1a), the gene encoding ZEB1, is induced by several potentially oncogenic ligands including TGF-beta, estrogen, and progesterone. TGF-beta appears to activate EMT, at least in part, by inducing ZEB1. However, our understanding of how ZEB1 contributes to signaling pathways elicited by estrogen and progesterone is quite limited, as is our understanding of its functional roles in normal adult tissues. To begin to address these questions, a human tissue mRNA array analysis was done. In adults, the highest ZEB1 mRNA expression is in bladder and uterus, whereas in the fetus highest expression is in lung, thymus, and heart. To further investigate the regulation of TCF8 by estrogen, ZEB1 mRNA was measured in ten estrogen-responsive cell lines, but it is only induced in the OV266 ovarian carcinoma line. Although high expression of ZEB1 mRNA is estrogen-dependent in normal human ovarian and endometrial biopsies, high expression is estrogen-independent in late stage ovarian and endometrial carcinomas, raising the possibility that deregulated expression promotes cancer progression. In contrast, TCF8 is at least partially deleted in 4 of 5 well-differentiated, grade I endometrial carcinomas, which may contribute to their non-aggressive phenotype. These data support the contention that high ZEB1 encourages gynecologic carcinoma progression.
Collapse
Affiliation(s)
- Elaine M Hurt
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD 21702, USA
| | | | | | | | | |
Collapse
|
17
|
Mao C, Patterson NM, Cherian MT, Aninye IO, Zhang C, Montoya JB, Cheng J, Putt KS, Hergenrother PJ, Wilson EM, Nardulli AM, Nordeen SK, Shapiro DJ. A new small molecule inhibitor of estrogen receptor alpha binding to estrogen response elements blocks estrogen-dependent growth of cancer cells. J Biol Chem 2008; 283:12819-30. [PMID: 18337247 PMCID: PMC2442351 DOI: 10.1074/jbc.m709936200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2007] [Revised: 03/12/2008] [Indexed: 12/21/2022] Open
Abstract
Estrogen receptor alpha (ERalpha) plays an important role in several human cancers. Most current ERalpha antagonists bind in the receptor ligand binding pocket and compete for binding with estrogenic ligands. Instead of the traditional approach of targeting estrogen binding to ER, we describe a strategy using a high throughput fluorescence anisotropy microplate assay to identify small molecule inhibitors of ERalpha binding to consensus estrogen response element (cERE) DNA. We identified small molecule inhibitors of ERalpha binding to the fluorescein-labeled (fl)cERE and evaluated their specificity, potency, and efficacy. One small molecule, theophylline, 8-[(benzylthio)methyl]-(7CI,8CI) (TPBM), inhibited ERalpha binding to the flcERE (IC(50) approximately 3 microm) and inhibited ERalpha-mediated transcription of a stably transfected ERE-containing reporter gene. Inhibition by TPBM was ER-specific, because progesterone and glucocorticoid receptor transcriptional activity were not significantly inhibited. In tamoxifen-resistant breast cancer cells that overexpress ERalpha, TPBM inhibited 17beta-estradiol (E(2))-ERalpha (IC(50) 9 microm) and 4-hydroxytamoxifen-ERalpha-mediated gene expression. Chromatin immunoprecipitation showed TPBM reduced E(2).ERalpha recruitment to an endogenous estrogen-responsive gene. TPBM inhibited E(2)-dependent growth of ERalpha-positive cancer cells (IC(50) of 5 microm). TPBM is not toxic to cells and does not affect estrogen-independent cell growth. TPBM acts outside of the ER ligand binding pocket, does not act by chelating the zinc in ER zinc fingers, and differs from known ERalpha inhibitors. Using a simple high throughput screen for inhibitors of ERalpha binding to the cERE, a small molecule inhibitor has been identified that selectively inhibits ERalpha-mediated gene expression and estrogen-dependent growth of cancer cells.
Collapse
Affiliation(s)
- Chengjian Mao
- Department of Biochemistry, and Chemistry, University of Illinois, Urbana, Illinois 61810-3602, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Pries R, Wittkopf N, Hasselbacher K, Wollenberg B. [Constitutive expression of the potential stem cell marker CD44 in permanent HNSCC cell lines]. HNO 2008; 56:461-6. [PMID: 18368378 DOI: 10.1007/s00106-008-1707-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Despite significant advances and the use of new diagnosic and therapy methods to treat head and neck squamous cell carcinoma (HNSCC), prognosis has improved only marginally in the last decades. Thus, there is an enormous need for novel immunotherapeutic approaches. It is becoming more and more obvious that stem cells play an important role in tumor development and progression. The identity of these cells and the underlying cellular and molecular mechanisms remain mostly unknown in HNSCC. MATERIAL AND METHODS Solid tumors as well as cells from the peripheral blood of patients with HNSCC were analyzed by flow cytometry concerning the expression of different putative stem cell marker proteins. RESULTS Distinct populations of CD44-positive (CD44+), lin-negative (lin-) potential stem cells could be identified in solid tumors of HNSCC patients with strong individual variations. Surprisingly, the potential stem cell marker CD44 was found to be constitutively expressed on the surface of all permanent HNSCC cell lines analyzed. CONCLUSION These data strongly suggest that CD44+ tumor stem cells may play a key role in establishment of permanent HNSCC cell lines, selecting especially robust cell entities that in real life might drive progression and metastasis of HNSCC. Individual analysis of tumor stem cell markers will be an important tool for innovative therapies and prognosis of patients with HNSCC. Additional stem cell markers will be investigated concerning their expression and cellular function.
Collapse
Affiliation(s)
- R Pries
- Klinik für HNO und Plastische Operationen, Universitätsklinikum Schleswig Holstein, Campus Lübeck
| | | | | | | |
Collapse
|
19
|
Salvo VA, Boué SM, Fonseca JP, Elliott S, Corbitt C, Collins-Burow BM, Curiel TJ, Srivastav SK, Shih BY, Carter-Wientjes C, Wood CE, Erhardt PW, Beckman BS, McLachlan JA, Cleveland TE, Burow ME. Antiestrogenic glyceollins suppress human breast and ovarian carcinoma tumorigenesis. Clin Cancer Res 2007; 12:7159-64. [PMID: 17145841 DOI: 10.1158/1078-0432.ccr-06-1426] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We have identified the phytoalexin compounds glyceollins I, II, and III, which exhibit marked antiestrogenic effects on estrogen receptor function and estrogen-dependent tumor growth in vivo. The purpose of this study was to investigate the interactions among the induced soy phytoalexins glyceollins I, II, and III on the growth of estrogen-dependent MCF-7 breast cancer and BG-1 ovarian cancer cells implanted in ovariectomized athymic mice. EXPERIMENTAL DESIGN Four treatment groups for each cell line were used: vehicle control, 20 mg/kg/mouse/d glyceollin mixture injection, 0.72 mg estradiol (E2) implant, and E2 implant + 20 mg/kg/mouse/d glyceollin injection. RESULTS Treatment with glyceollin suppressed E2-stimulated tumor growth of MCF-7 cells (-53.4%) and BG-1 cells (-73.1%) in ovariectomized athymic mice. These tumor-inhibiting effects corresponded with significantly lower E2-induced progesterone receptor expression in the tumors. In contrast to tamoxifen, the glyceollins had no estrogen-agonist effects on uterine morphology and partially antagonized the uterotropic effects of estrogen. CONCLUSIONS These findings identify glyceollins as antiestrogenic agents that may be useful in the prevention or treatment of breast and ovarian carcinoma.
Collapse
Affiliation(s)
- Virgilo A Salvo
- Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zhou R, Treeck O, Horn F, Ortmann O. Effects of prolonged tamoxifen treatment on receptor expression and apoptosis of ovarian cancer cells. Gynecol Oncol 2005; 96:678-83. [PMID: 15721411 DOI: 10.1016/j.ygyno.2004.11.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2004] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Tamoxifen, which is widely used in the treatment of breast cancer, also has a beneficial effect on cisplatin-refractory ovarian cancer. In this study, we investigated the long-term effects of this drug on estrogen-receptor-positive ovarian cancer cells. METHODS We performed an in vitro selection process by long-term treatment of BG-1 ovarian cancer cells with 4-hydroxy tamoxifen (4-OH TAM). Drug effects on cell growth were determined by measurement of relative cell numbers (MTS assay), the apoptotic effects of 4-OH TAM were determined by analysis of poly (ADP-ribose) polymerase (PARP) cleavage and by ELISA measurement of DNA-histone complexes in cytoplasm. RESULTS Analysis of BG-1(LT) ovarian cancer cells isolated after 5 months of long-term treatment with 4-OH TAM revealed both a significantly reduced apoptotic and antiproliferative effect of this drug. Further experiments to examine expression changes of the receptor tyrosine kinases EGFR, HER2 and estrogen receptor alpha did not reveal any alterations in BG-1(LT) if compared to wild-type cells. In contrast, in this cell line, a significant alteration in the expression of estrogen receptor beta was observed. CONCLUSION Our findings indicate that long-term treatment with 4-OH TAM is able to diminish both the antiproliferative and apoptotic action of this drug on BG-1 ovarian cancer cells. Our data suggest that the responsiveness of ovarian cancer cells to 4-OH TAM decreases after long-term treatment with this drug in vitro like previously observed after long-term treatment of breast cancer cells.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, 610041 Chengdu, PR China
| | | | | | | |
Collapse
|
21
|
Bardin A, Moll F, Margueron R, Delfour C, Chu ML, Maudelonde T, Cavailles V, Pujol P. Transcriptional and posttranscriptional regulation of fibulin-1 by estrogens leads to differential induction of messenger ribonucleic acid variants in ovarian and breast cancer cells. Endocrinology 2005; 146:760-8. [PMID: 15528301 DOI: 10.1210/en.2004-1239] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Fibulin-1 is an extracellular matrix protein overexpressed in epithelial ovarian and breast cancers. In estrogen receptor (ER)-positive ovarian and breast cancer cell lines, fibulin-1 mRNA levels are markedly increased by estrogens. Transfection experiments using fibulin-1 promoter constructs indicate that 17beta-estradiol (E2) increases fibulin-1 gene transcription and that ERalpha is more potent than ERbeta to mediate E2 regulation of the transfected fibulin-1 promoter. Using SL2 cells devoid of specificity protein 1 (Sp1) and site-directed mutagenesis of GC boxes, we evidenced that the E2 regulation occurs through a proximal specificity protein 1 binding site. In addition, we show that fibulin-1C and -1D mRNAs, the two major fibulin-1 splicing variants, are differentially induced by E2. The induction of both mRNAs variants is direct and independent of a newly synthesized protein intermediate. Interestingly, actinomycin D chase experiments demonstrate that E2 treatment selectively shortens the fibulin-1D mRNA half-life. This indicates that estrogens affect differentially the stability of fibulin-1 variants and may explain the lower accumulation of fibulin-1D mRNA on E2 treatment. In conclusion, our data show that estrogens, via ERalpha, are key regulators of fibulin-1 expression at both the transcriptional and posttranscriptional levels. The preferential induction of the fibulin-1C variant, which is overexpressed in ovarian and breast cancer, might play an important role in estrogen-promoted carcinogenesis.
Collapse
Affiliation(s)
- A Bardin
- Unité Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Montpellier, Hôpital Arnaud de Villeneuve, 34095 Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Kalli KR, Bradley SV, Fuchshuber S, Conover CA. Estrogen receptor-positive human epithelial ovarian carcinoma cells respond to the antitumor drug suramin with increased proliferation: possible insight into ER and epidermal growth factor signaling interactions in ovarian cancer. Gynecol Oncol 2004; 94:705-12. [PMID: 15350362 DOI: 10.1016/j.ygyno.2004.05.059] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2004] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Development of targeted therapeutics for ovarian cancer requires a basic understanding of ovarian epithelial carcinoma cell biology. The role of estrogen and epidermal growth factor (EGF) in control of cell growth was investigated in a panel of ovarian carcinoma lines. METHODS EGF receptor (EGFR) was detected by flow cytometry and estrogen receptor (ER) by Northern blot. Western blotting and [(3)H]thymidine incorporation were used to determine receptor activation and the effects of ligand exposure and growth factor antagonists, including the antineoplastic agent, suramin, on cell growth. RESULTS Only one cell line, OV266, expressed ER and responded to beta-estradiol with increases in DNA synthesis and cell proliferation that could be blocked by the pure antiestrogen ICI 182,780. All cell lines possessed functional EGFR as measured by flow cytometry and phosphorylation of the receptor and mitogen-activated protein kinase after EGF treatment, but only two cell lines (OV177 and OV266) proliferated in response to exogenous EGF. Suramin had limited effectiveness in inhibiting growth in four of five cell lines and had a striking dose-dependent stimulatory effect on OV266 cell growth. The proliferative response to suramin could be inhibited with EGFR antagonists. CONCLUSION Cultured epithelial ovarian carcinoma cell lines express EGFR (5/5) and can express ER (1/5). Differential growth responses to EGF were observed despite uniform receptor and MAPK activation. Unexpectedly, the antineoplastic agent suramin increased growth of ER positive ovarian carcinoma cells in an EGFR-dependent manner. These studies provide insight into the complex interactions of these systems in control of ovarian cancer cell growth.
Collapse
Affiliation(s)
- Kimberly R Kalli
- Department of Oncology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|
23
|
Giacalone PL, Daurés JP, Ouafik L, Martin PM, Laffargue F, Maudelonde T. Steroids and adrenomedullin growth patterns in human ovarian cancer cells: estrogenic-regulation assay. Gynecol Oncol 2003; 91:651-6. [PMID: 14675694 DOI: 10.1016/s0090-8258(03)00279-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Adrenomedullin is a multifunctional regulatory peptide with mitogenic and angiogenic capabilities. Objectives in this study were: (1) to describe the effects of adrenomedullin and anti-adrenomedullin antibodies on ovarian carcinoma cell proliferation, and (2) to assess the estradiol regulation of adrenomedullin metabolism. METHODS We assessed the effects of estradiol, adrenomedullin, and anti-adrenomedullin antibodies on cell growth in three human ovarian cell lines. RT-PCR was used to assess mRNA expression and Western blots to determine protein levels. RESULTS Estradiol stimulates BG-1 and PEO4 cells growth but not PEO14 cells. Adrenomedullin mRNA expression and secretion were not under estrogen control. Adrenomedullin and anti-adrenomedullin antibodies had no growth effects in vitro. Adjunction of anti-adrenomedullin antibodies to estradiol-stimulated cells significantly inhibited their growth. CONCLUSION Adrenomedullin metabolism is not under estradiol control. Anti-adrenomedullin antibodies display inhibitory effects on cells having high mitogenic activity. This opens the need for additional search toward in vivo specific immunotherapy.
Collapse
Affiliation(s)
- Pierre-Ludovic Giacalone
- Department of Obstetrics and Gynecology (Pr F Laffargue), Hôpital Arnaud de Villeneuve, University of Montpellier, Montpellier, France.
| | | | | | | | | | | |
Collapse
|
24
|
Hamilton TC, Connolly DC, Nikitin AY, Garson K, Vanderhyden BC. Translational research in ovarian cancer: a must. Int J Gynecol Cancer 2003; 13 Suppl 2:220-30. [PMID: 14656284 DOI: 10.1111/j.1525-1438.2003.13350.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Ovarian cancer discovered at late clinical stage continues to be a fatal disease. It seems self-evident that if we are to make an impact on the survival of advanced ovarian cancer patients, we must begin to understand the disease more completely. This should improve the diagnosis of the disease at an early stage when it is curable by surgery or develop better/targeted drug treatments. Modern molecular techniques have provided insights into many of the molecular changes that occur when ovarian cancer develops, but one must understand that changes seen in this way can only be said to correlate with disease. It would be helpful to have a way to test candidate changes for causality. In many cancer types, genetically engineered animals are beginning to be used for this purpose and as a means to study the disease process in greater detail. To date, there has been no way to study ovarian cancer by this means. Efforts to model human ovarian cancer have been delayed by a general lack of understanding both of the disease process in humans and of the cells widely believed to be the precursors of epithelial ovarian cancer, the ovarian surface epithelial (OSE) cells. Here, we present recent progress in modeling ovarian cancer using genetically modified mice.
Collapse
Affiliation(s)
- T C Hamilton
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | | | | | | |
Collapse
|
25
|
Luo LY, Grass L, Diamandis EP. Steroid hormone regulation of the human kallikrein 10 (KLK10) gene in cancer cell lines and functional characterization of the KLK10 gene promoter. Clin Chim Acta 2003; 337:115-26. [PMID: 14568187 DOI: 10.1016/j.cccn.2003.07.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND The human kallikrein 10 (KLK10) gene is a new member of the human tissue kallikrein gene family. It encodes for a secreted serine protease (hK10) with predicted trypsin-like enzymatic activity. KLK10 is highly expressed in the sex organs and its expression level changes in malignancy. METHODS To determine the role of steroid hormones in KLK10 gene expression, we investigated its modulation by 17beta-estradiol, 5alpha-dihydrotestosterone, norgestrel, dexamethasone and aldosterone, at both the transcription and translation level, in a panel of cancer cell lines. After steroid hormone stimulation, the change of KLK10 mRNA was monitored with reverse transcriptase polymerase chain reaction and hK10 protein levels in the culture supernatant were quantified with an hK10-specific immunoassay. The presence of hormone response elements in the KLK10 gene promoter was examined with the chloramphenicol acetyltransferase reporter gene system. RESULTS The KLK10 expression was mainly up-regulated by estrogens, androgens and progestins, and to a lesser extent by dexamethasone and aldosterone in the breast cancer cell lines BT-474, MCF-7 and T-47D, both at the mRNA and protein levels. The effect of stimulation of these steroids on KLK10 expression varied among the cell lines. Estrogens, androgens and progestins were most potent in the BT-474, T-47D and MCF-7 cells, respectively. The up-regulation effect of estrogens, androgens, and progestins on KLK10 expression can be blocked by their antagonists ICI-182, 780, RU-56,187, and mifepristone, respectively. Time course studies showed that hK10 protein started to increase 1 day after steroid hormone stimulation and this increase persisted for 7 days. These data suggest that steroid hormones up-regulate KLK10 gene expression through direct interaction between hormone-receptor complexes and their cognate hormone response elements. To search for hormone response elements, we functionally characterized the KLK10 promoter by placing it upstream of the chloramphenicol acetyltransferase reporter gene. We found that KLK10 promoter activity did not rely on the presence of functional estrogen and androgen receptors. Also, the presence of functional estrogen and androgen receptors did not increase its constitutive activity. We suggest that the hormone response elements that mediate the transcriptional regulation of KLK10 are unlikely to locate in the KLK10 promoter. CONCLUSIONS Estrogens, androgens and progestins modulate KLK10 expression through their own receptors but this regulation is not mediated by steroid hormone response elements in the promoter of the KLK10 gene.
Collapse
Affiliation(s)
- Liu-Ying Luo
- Department of Pathology, Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5
| | | | | |
Collapse
|
26
|
Nicosia SV, Bai W, Cheng JQ, Coppola D, Kruk PA. Oncogenic pathways implicated in ovarian epithelial cancer. Hematol Oncol Clin North Am 2003; 17:927-43. [PMID: 12959183 DOI: 10.1016/s0889-8588(03)00056-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Characterization of intracellular signaling pathways should lead to a better understanding of ovarian epithelial carcinogenesis and provide an opportunity to interfere with signal transduction targets involved in ovarian tumor cell growth, survival, and progression. Challenges toward such an effort are significant because many of these signals are part of cascades within an intricate and likely redundant intracellular signaling network (Fig.1). For instance, a given signal may activate a dual intracellular pathway (ie, MEK1-MAPK and PI3K/Akt required for fibronectin-dependent activation of matrix metalloproteinase 9). A single pathway also may transduce more than one biologic or oncogenic signal (ie, PI3K signaling in epithelial and endothelial cell growth and sprouting of neovessels). Despite these challenges, evidence for therapeutic targeting of signal transduction pathways is accumulating in human cancer. For instance, the EGF-specific tyrosine kinase inhibitor ZD 1839 (Iressa) may have a beneficial therapeutic effect on ovarian epithelial cancer. Therapy of this cancer may include inhibitors of PI kinase (quercetin), ezrin and PIP kinase (genistein). The G protein-coupled family of receptors, including LPA, also is an attractive target to drugs, although their frequent pleiotropic functions may be at times toxic and lack specificity. Because of the lack of notable toxicity, PI3K/Akt pathway inhibitors such as FTIs are a promising targeted therapy of ovarian epithelial cancer. Increasing insight into the oncogenic pathways involved in ovarian epithelial cancer also is helping clinicians to understand better the phenomenon of chemoresistance in this malignancy. Oncogenic activation of gamma-synuclein promotes cell survival and provides resistance to paclitaxel, but such a resistance is partially overcome by an MEK inhibitor that suppresses ERK activity. Ovarian epithelial cancer is a complex group of neoplasms with an overall poor prognosis. Comprehension of this cancer pathobiology suffers because of an incomplete understanding of precursor lesions and the absence of an orthotopic animal model until very recently. It can be predicted with confidence, however, that the discovery of potent inhibitors of signal transduction and the development of discovery tools, such as proteomics and metabolomics, may change the way by which clinicians may now address basic biomedical questions in this insidious and lethal disease.
Collapse
Affiliation(s)
- Santo V Nicosia
- Department of Pathology and Laboratory Medicine, University of South Florida College of Medicine, 12901 Bruce B. Downs Boulevard, MDC Box 11, Tampa, FL 33612, USA.
| | | | | | | | | |
Collapse
|
27
|
Jiang F, Saunders BO, Haller E, Livingston S, Nicosia SV, Bai W. Conditionally immortal ovarian cell lines for investigating the influence of ovarian stroma on the estrogen sensitivity and tumorigenicity of ovarian surface epithelial cells. In Vitro Cell Dev Biol Anim 2003; 39:304-12. [PMID: 12964904 DOI: 10.1290/1543-706x(2003)039<0304:cioclf>2.0.co;2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The tendency of the ovarian surface epithelium (OSE) to undergo metaplastic and morphogenetic changes during the life cycle, at variance with the adjacent peritoneal mesothelial cells, suggests that its biology may be regulated by underlying ovarian stromal cues. However, little is known about the role that the ovarian stroma plays in the pathobiology of the OSE, largely because of the lack of a suitable in vitro model. Here, we describe the establishment and characterization of conditionally immortalized ovarian stromal and surface epithelial cell lines from H-2K(b)-tsA58 transgenic mice that carry the thermolabile mutant of SV-40 large T antigen under the control of an interferon-gamma (IFN-gamma)-inducible promoter. These cells express functional T antigens, grow continuously under permissive conditions at 33 degrees C in the presence of IFN-gamma, and stop dividing when the activity and expression of the tumor antigen is suppressed by restrictive conditions without IFN-gamma at 39 degrees C. Morphological, immunohistochemical, and ultrastructural analyses show that conditionally immortal OSE cells form cobblestone-like monolayers, express cytokeratin and vimentin, contain several microvilli, and develop tight junctions, whereas stromal cells are spindle-like, express vimentin but not cytokeratin, and contain rare microvilli, thus exhibiting epithelial and stromal phenotypes, respectively. At variance with the reported behavior of rat epithelial cells, conditionally immortal mouse epithelial cells are not spontaneously transformed after continuous culture in vitro. More importantly, conditioned media from stromal cells cultured under permissive conditions increase the specific activity of the endogenous estrogen receptor in BG-1 human ovarian epithelial cancer cells and promote these cells' anchorage-independent growth, suggesting the paracrine influence of a stromal factor. In addition, stromal cells cultured under restrictive conditions retain this growth-stimulatory activity, which, therefore, appears to be independent of T antigen expression. These established cell lines should provide a useful in vitro model system for studying the role of cellular interactions in OSE cell growth and tumorigenesis.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Pathology, University of South Florida College of Medicine and H. Lee Moffitt Cancer Center, Tampa, Florida 33612-4799, USA
| | | | | | | | | | | |
Collapse
|
28
|
Srinivas G, Annab LA, Gopinath G, Banerji A, Srinivas P. Antisense blocking of BRCA1 enhances sensitivity to plumbagin but not tamoxifen in BG-1 ovarian cancer cells. Mol Carcinog 2003; 39:15-25. [PMID: 14694444 DOI: 10.1002/mc.10164] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Previous studies have shown that reduction in BRCA1 mRNA and protein can result in increased proliferation of BG-1 ovarian cancer cells in both in vitro and in vivo conditions, suggesting that BRCA1 may normally act as a growth inhibitor in these cells. Also, there are other reports that suggest that wild-type BRCA1 protein may repress estrogen receptor (ER) function either directly or indirectly. However, response to antiestrogen drugs in BRCA1-blocked ER-positive ovarian cancer cells has not been reported, and this served as the rationale for this study. We analyzed the effect of tamoxifen, emodin, and plumbagin in BRCA1-blocked ER-positive BG-1 ovarian cancer cells. For all three drugs, BRCA1-blocked cells were more sensitive than the corresponding control cells as assessed by MTT assay; however, only plumbagin showed a statistically significant difference in mean viability (P < 0.05). All three drugs induced loss of mitochondrial membrane potential (DeltaPsi(m)), nuclear condensation, DNA fragmentation, and morphological changes, as observed after 6 h of drug treatment, suggesting apoptosis induction in both BRCA1-blocked and control cells. However, apoptosis induction was greater in BRCA1-blocked cells, the efficacy being in the order of plumbagin > tamoxifen > emodin. The dose of plumbagin needed to kill 50% was 5 microM in the control cells and 2.68 microM for the BRCA1-blocked cells, indicating that the latter was about twofold more sensitive to plumbagin than the wild-type cells. This throws light on the fact that plumbagin may have chemotherapeutic potential as an anticancer agent in BRCA1-mutated ovarian cancer patients.
Collapse
Affiliation(s)
- Gopal Srinivas
- Division of Cancer Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | | | | | | | | |
Collapse
|
29
|
|
30
|
Schmitt E, Dekant W, Stopper H. Assaying the estrogenicity of phytoestrogens in cells of different estrogen sensitive tissues. Toxicol In Vitro 2001; 15:433-9. [PMID: 11566575 DOI: 10.1016/s0887-2333(01)00048-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
There is currently much concern that a wide range of both synthetic and naturally occurring environmental chemicals may act as endocrine disruptors (ED), and may adversely affect humans and wildlife. We examined the estrogenic effects of the phytoestrogens daidzein (DAI), equol (EQU) and O-desmethylangolensin (O-DMA), two metabolites of DAI, in three different assays. Binding affinity to the estrogen receptor alpha was 1000-10,000-fold lower compared with the endogenous estrogen estradiol. In the receptor positive cell line MCF-7 the phytoestrogens induced the expression of a reporter gene. The E-SCREEN is based on the estrogen-receptor binding induced proliferation of the human breast cancer cell line MCF-7. We also adapted the E-SCREEN for the estrogen-receptor positive human ovarian cancer cell line BG-1. The tested phytoestrogens induced cell proliferation in both cell lines, but not in the receptor negative human breast cancer cell line MDA-MB-231. The phytoestrogen-induced cell proliferation could be blocked by addition of the receptor antagonist 4-hydroxytamoxifen (OHT). Combination treatments with the endogenous estrogen estradiol showed competitive effects in MCF-7 cells. These studies demonstrated that the tested phytoestrogens exerted estrogenic responses in cells derived from two different tissues, breast and ovary. Furthermore, we demonstrated that BG-1 cells are a suitable additional cell system to investigate estrogenicity of test compounds.
Collapse
Affiliation(s)
- E Schmitt
- Department of Toxicology, University of Wuerzburg, Versbacherstrasse 9, D-97078 Wuerzburg, Germany.
| | | | | |
Collapse
|
31
|
Annab LA, Hawkins RE, Solomon G, Barrett JC, Afshari CA. Increased cell survival by inhibition of BRCA1 using an antisense approach in an estrogen responsive ovarian carcinoma cell line. Breast Cancer Res 2000; 2:139-48. [PMID: 11056686 PMCID: PMC13916 DOI: 10.1186/bcr45] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/1999] [Revised: 12/07/1999] [Accepted: 01/20/2000] [Indexed: 11/10/2022] Open
Abstract
STATEMENT OF FINDINGS: We tested the hypothesis that BRCA1 may play a role in the regulation of ovarian tumor cell death as well as the inhibition of ovarian cell proliferation. Introduction of BRCA1 antisense retroviral constructs into BG-1 estrogen-dependent ovarian adenocarcinoma cells resulted in reduced BRCA1 expression. BRCA1 antisense pooled populations and derived subclones were able to proliferate in monolayer culture without estrogen, whereas control cells began to die after 10 days of estrogen deprivation. In addition, both populations and subclones of BRCA1 antisense infected cells demonstrated a growth advantage in monolayer culture in the presence of estrogen and were able to proliferate in monolayer culture without estrogen, while control cells did not. Furthermore, clonal studies demonstrated that reduced levels of BRCA1 protein correlated with growth in soft agar and greater tumor formation in nude mice in the absence of estrogen. These data suggest that reduction of BRCA1 protein in BG-1 ovarian adenocarcinoma cells may have an effect on cell survival during estrogen deprivation both in vitro and in vivo.
Collapse
Affiliation(s)
- L A Annab
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | | | | | | | | |
Collapse
|