1
|
DOLIN A, SCHWEIGER P, WASELAU M, EGERBACHER M, WALTER I. Immunohistochemical markers for equine granulosa cell tumors: a pilot study. J Equine Sci 2023; 34:37-46. [PMID: 37405069 PMCID: PMC10315638 DOI: 10.1294/jes.34.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 01/18/2023] [Indexed: 07/06/2023] Open
Abstract
Sex cord-stromal tumors (SCSTs), generally referred to as granulosa cell tumors (GCTs) or granulosa-theca cell tumors (GTCTs) in equids, show complex compositions and variable numbers of hormone-producing cells. These tumors can be difficult to diagnose, especially in early stages. Therefore, we tested a panel of antibodies for vimentin, smooth muscle actin, laminin, Ki-67, E-cadherin, calretinin, moesin, p-ezrin, AMH, and aromatase, markers used for tumor composition and classification, progression, and prognosis in human SCSTs, on an exemplary grapefruit-size equine GCT within the left ovary of a 13-year-old mare with stallion-like behavior and elevated testosterone levels in comparison with normal ovarian tissue. The tumor showed a low proliferation rate and prominent moesin and p-ezrin staining in granulosa cells. E-cadherin, calretinin, aromatase, and AMH are suggested to be potential markers for different cell components of equine SCSTs that can support tumor diagnosis and classification.
Collapse
Affiliation(s)
| | | | - Martin WASELAU
- Equine Hospital Aschheim, Equine Diagnostic
Center Munich, 85609 Munich, Germany
| | - Monika EGERBACHER
- Administrative Unit Veterinary Medicine, UMIT
TIROL-Private University for Health Sciences, Medical Informatics and Technology GmbH,
6060 Hall in Tirol, Austria
| | - Ingrid WALTER
- Department of Pathobiology, Institute of
Morphology, Vetmeduni Vienna, 1210 Vienna, Austria
| |
Collapse
|
2
|
Tossetta G, Fantone S, Goteri G, Giannubilo SR, Ciavattini A, Marzioni D. The Role of NQO1 in Ovarian Cancer. Int J Mol Sci 2023; 24:ijms24097839. [PMID: 37175546 PMCID: PMC10178676 DOI: 10.3390/ijms24097839] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Ovarian cancer is one of the most dangerous gynecologic malignancies showing a high fatality rate because of late diagnosis and relapse occurrence due to chemoresistance onset. Several researchers reported that oxidative stress plays a key role in ovarian cancer occurrence, growth and development. The NAD(P)H:quinone oxidoreductase 1 (NQO1) is an antioxidant enzyme that, using NADH or NADPH as substrates to reduce quinones to hydroquinones, avoids the formation of the highly reactive semiquinones, then protecting cells against oxidative stress. In this review, we report evidence from the literature describing the effect of NQO1 on ovarian cancer onset and progression.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Gaia Goteri
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, 60126 Ancona, Italy
| | | | - Andrea Ciavattini
- Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, 60123 Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| |
Collapse
|
3
|
Yue H, Yang X, Wu X, Tian Y, Xu P, Sang N. Identification of risk for ovarian disease enhanced by BPB or BPAF exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 319:120980. [PMID: 36587784 DOI: 10.1016/j.envpol.2022.120980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/06/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
The ban on bisphenol A (BPA) has led to a rapid increase in the use of BPA analogs, and they are increasingly being detected in the natural environment and biological organisms. Studies have pointed out that BPA analogs can lead to adverse health outcomes. However, their interference with ovarian tissue has not been fully elucidated. In this study, seven- to eight-week-old CD-1 mice were exposed to corn oil containing 300 μg/kg/day bisphenol B (BPB) or bisphenol AF (BPAF) through oral gavage, and ovarian tissues were collected at 14 and 28 days of exposure. Ovarian toxicity was evaluated by the ovarian index, ovarian area, and follicle number. mRNA-seq was used to identify differentially expressed genes (DEGs) and infer the association of DEGs with ovarian diseases. BPB or BPAF exposure induced morphological changes in ovarian tissue in CD-1 mice. In addition, Gene Ontology (GO) analysis revealed disturbances in biological processes (BP) associated with steroid biosynthetic process (GO:0006694) and cellular calcium ion homeostasis (GO:0006874). Subsequently, regulatory networks of BPA analogs (BPB or BPAF)-DEGs-ovarian diseases were constructed. Importantly, the expression levels of DEGs and transcription factors (TFs) associated with ovarian disease were altered. BPB or BPAF exposure causes damage to ovarian morphology through the synergistic effects of multiple biological processes and may be associated with altered mRNA expression profiles as a risk factor for ovarian diseases.
Collapse
Affiliation(s)
- Huifeng Yue
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| | - Xiaowen Yang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Xiaoyun Wu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Yuchai Tian
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Pengchong Xu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| |
Collapse
|
4
|
Sun X, Tang F, Guo Q, Liu Y, He Y, Du Y, Gao F, Zhang G, Yang C. HAS2-Ezrin-ER axis plays a role in acquired antiestrogen resistance of ER-positive breast cancer. Front Pharmacol 2022; 13:1031487. [PMID: 36386154 PMCID: PMC9659586 DOI: 10.3389/fphar.2022.1031487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
The development of endocrine resistance is a major clinical problem in estrogen receptor-positive (ER+) breast cancer (BrCa) treatment, in which how cancer cells acquire resistance remains obscure. Hyaluronan synthase 2 (HAS2) is the most critical synthase in producing hyaluronan and is well known for its involvement in cancer growth, metabolism and metastasis. Recent evidence has proved that HAS2 is involved in cellular acquired resistance to drug therapy in BrCa. In this work, we first observed that HAS2 expression was decreased in the endocrine-resistant ER+ BrCa cells. Further knocking-out experiments confirmed that the loss of HAS2 in parental ER+ BrCa cells resulted in a following antiestrogen resistance. Next, we found that the HAS2-loss could induce an upregulation of Ezrin, a member of the membrane cytoskeletal protein family who plays key roles in cellular signal transduction. Notably, we identified that the increase of Ezrin induced by HAS2-loss could inhibit the ERα expression and augment antiestrogen resistance, suggesting that a HAS2-Ezrin-ER axis may be associated with the acquirement of endocrine resistance in ER+ BrCa cells. Finally, knockdown or inhibition of Ezrin could restore the sensitivity of endocrine-resistant cells to antiestrogens treatment by activating ERα signaling. Taken together, our findings unraveled a novel HAS2-Ezrin-ER route in regulating the sensitivity of ER+ BrCa cells to antiestrogens, in which Ezrin may be a potential target in endocrine therapy.
Collapse
Affiliation(s)
- Xiaodan Sun
- Department of Clinical Laboratory, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fen Tang
- Department of Breast Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Guo
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Liu
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqing He
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Du
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Gao
- Department of Clinical Laboratory, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoliang Zhang
- Department of Clinical Laboratory, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cuixia Yang
- Department of Clinical Laboratory, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Molecular Biology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Pavlič R, Gjorgoska M, Rižner TL. Model Cell Lines and Tissues of Different HGSOC Subtypes Differ in Local Estrogen Biosynthesis. Cancers (Basel) 2022; 14:cancers14112583. [PMID: 35681563 PMCID: PMC9179372 DOI: 10.3390/cancers14112583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Ovarian cancer (OC) comprises a heterogeneous group of hormone-dependent diseases with very high mortality. Estrogens have been shown to promote the progression of OC; however, their exact role in OC subtypes remains unknown. Here, we investigated the local estrogen biosynthesis in OC. We performed targeted transcriptomics and estrogen metabolism analyses in high-grade serous OC (HGSOC) cell lines that differed in chemoresistance status and compared these data with publicly available transcriptome and proteome data for HGSOC tissues. In HGSOC cells, estrogen metabolism decreased with increasing chemoresistance. In highly chemoresistant cells and platinum-resistant HGSOC tissues, HSD17B14 expression was increased. Proteome data showed differential levels of HSD17B10, SULT1E1, CYP1B1, and NQO1 between the four HGSOC subtypes. Our results confirm that estrogen biosynthesis differs between different HGSOC cell models and possibly between different HGSOC subtypes. Such differentially expressed enzymes have potential as targets in the search of new treatment options. Abstract Ovarian cancer (OC) is highly lethal and heterogeneous. Several hormones are involved in OC etiology including estrogens; however, their role in OC is not completely understood. Here, we performed targeted transcriptomics and estrogen metabolism analyses in high-grade serous OC (HGSOC), OVSAHO, Kuramochi, COV632, and immortalized normal ovarian epithelial HIO-80 cells. We compared these data with public transcriptome and proteome data for the HGSOC tissues. In all model systems, high steroid sulfatase expression and weak/undetected aromatase (CYP19A1) expression indicated the formation of estrogens from the precursor estrone-sulfate (E1-S). In OC cells, the metabolism of E1-S to estradiol was the highest in OVSAHO, followed by Kuramochi and COV362 cells, and decreased with increasing chemoresistance. In addition, higher HSD17B14 and CYP1A2 expressions were observed in highly chemoresistant COV362 cells and platinum-resistant tissues compared to those in HIO-80 cells and platinum-sensitive tissues. The HGSOC cell models differed in HSD17B10, CYP1B1, and NQO1 expression. Proteomic data also showed different levels of HSD17B10, CYP1B1, NQO1, and SULT1E1 between the four HGSOC subtypes. These results suggest that different HGSOC subtypes form different levels of estrogens and their metabolites and that the estrogen-biosynthesis-associated targets should be further studied for the development of personalized treatment.
Collapse
|
6
|
Tameishi M, Ishikawa H, Tanaka C, Kobori T, Urashima Y, Ito T, Obata T. Ezrin Contributes to the Plasma Membrane Expression of PD-L1 in A2780 Cells. J Clin Med 2022; 11:jcm11092457. [PMID: 35566582 PMCID: PMC9100183 DOI: 10.3390/jcm11092457] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 01/30/2023] Open
Abstract
Programmed death ligand–1 (PD–L1) is one of the immune checkpoint molecule localized on the plasma membrane of numerous cancer cells that negatively regulates T-cell-mediated immunosurveillance. Despite the remarkable efficacy and safety profile of immune checkpoint inhibitors (ICIs), such as anti-PD–L1 antibodies, restricted poor therapeutic responses to ICIs are often observed in patients with ovarian cancer. Because higher expression of PD–L1 in advanced ovarian cancer is associated with a decreased survival rate, identifying the potential molecules to regulate the plasma membrane expression of PD–L1 may provide a novel therapeutic strategy to improve the efficacy of ICIs against ovarian cancers. Here, we reveal the involvement of the ezrin/radixin/moesin (ERM) family, which crosslinks transmembrane proteins with the actin cytoskeleton by serving as a scaffold protein, in the plasma membrane expression of PD–L1 in the human epithelial ovarian cancer cell line A2780. Our results demonstrate that PD–L1 and all three ERMs were expressed at the mRNA and protein levels in A2780 cells, and that PD–L1 was highly colocalized with ezrin and moesin, but moderately with radixin, in the plasma membrane. Interestingly, RNA interference-mediated gene silencing of ezrin, but not of radixin or moesin, substantially reduced the plasma membrane expression of PD–L1 without altering its mRNA expression. In conclusion, our results indicate that ezrin may be responsible for the plasma membrane expression of PD–L1, possibly by serving as a scaffold protein in A2780 cells. Ezrin is a potential therapeutic target for improving the efficacy of ICIs against ovarian cancers.
Collapse
Affiliation(s)
- Mayuka Tameishi
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Honami Ishikawa
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Chihiro Tanaka
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Takuro Kobori
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Yoko Urashima
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
| | - Takuya Ito
- Laboratory of Natural Medicines, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan;
| | - Tokio Obata
- Laboratory of Clinical Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi 584-8540, Japan; (M.T.); (H.I.); (C.T.); (T.K.); (Y.U.)
- Correspondence: ; Tel.: +81-721-24-9371
| |
Collapse
|
7
|
Tuesley KM, Webb PM, Protani MM, Spilsbury K, Pearson SA, Coory MD, Donovan P, Steer C, Stewart LM, Pandeya N, Jordan SJ. Nitrogen-Based Bisphosphonate Use and Ovarian Cancer Risk in Women Aged 50 Years and Older. J Natl Cancer Inst 2022; 114:878-884. [PMID: 35262727 PMCID: PMC9194625 DOI: 10.1093/jnci/djac050] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/23/2022] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND There are few readily modifiable risk factors for epithelial ovarian cancer; pre-clinical studies suggest bisphosphonates could have chemo-preventive actions. Our study aimed to assess the association between use of nitrogen-based bisphosphonate medicine and risk of epithelial ovarian cancer, overall and by histotype. METHODS We conducted a case-control study nested within a large linked administrative dataset including all Australian women enrolled for Medicare, Australia's universal health insurance scheme, between July 2002 and December 2013. We included all women with epithelial ovarian cancer diagnosed at age 50 years and older between 1st July 2004 and 31st December 2013 (n = 9,367) and randomly selected up to five controls per case, individually matched to cases by age, state of residence, area-level socioeconomic status, and remoteness of residence category (n = 46,830). We used prescription records to ascertain use of nitrogen-based bisphosphonates (ever use and duration of use), raloxifene and other osteoporosis medicines (non-nitrogen-based bisphosphonates, strontium and denosumab). We calculated adjusted odds ratios (OR) and 95% confidence intervals (CI) using conditional logistic regression. RESULTS Ever use of nitrogen-based bisphosphonates was associated with a reduced risk of epithelial ovarian cancer compared to non-use (OR = 0.81, 95%CI : 0.75-0.88). There was a reduced risk of both endometrioid (OR = 0.51, 95%CI : 0.33-0.79) and serous histotypes (OR = 0.84, 95%CI : 0.75-0.93), but no association with the mucinous or clear cell histotypes. CONCLUSION Use of nitrogen-based bisphosphonates was associated with a reduced risk of endometrioid and serous ovarian cancer. This suggests the potential for use for prevention, although validation of our findings is required.
Collapse
Affiliation(s)
- Karen M Tuesley
- School of Public Health, Faculty of Medicine, University of Queensland, Brisbane, Australia.,Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Penelope M Webb
- School of Public Health, Faculty of Medicine, University of Queensland, Brisbane, Australia.,Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Melinda M Protani
- School of Public Health, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Katrina Spilsbury
- Institute for Health Research, The University of Notre Dame Australia, Fremantle, Australia
| | | | - Michael D Coory
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Peter Donovan
- Clinical Pharmacology Department, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Christopher Steer
- Border Medical Oncology, Albury-Wodonga Regional Cancer Centre, Albury, Australia.,University of NSW Rural Clinical School, Albury Campus, Albury, New South Wales, Australia
| | - Louise M Stewart
- School of Population and Global Health, The University of Western Australia, Perth, Australia
| | - Nirmala Pandeya
- School of Public Health, Faculty of Medicine, University of Queensland, Brisbane, Australia.,Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Susan J Jordan
- School of Public Health, Faculty of Medicine, University of Queensland, Brisbane, Australia.,Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
8
|
Lv X, Miao C, Liu M, Wang X, Wang L, Wang D. 17β-Estradiol via Orai1 activates calcium mobilization to induce cell proliferation in epithelial ovarian cancer. J Biochem Mol Toxicol 2020; 34:e22603. [PMID: 32844545 DOI: 10.1002/jbt.22603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/21/2020] [Accepted: 08/10/2020] [Indexed: 11/06/2022]
Abstract
Epithelial ovarian cancer (EOC) is the most lethal estrogen-sensitive gynecological cancer. Studies have reported that estrogen induces rapid cellular calcium mobilization in cells and can determine the fate of a cell. We found that estrogen increased the calcium release-activated calcium channel modulator 1 (Orai1) protein expression levels in SK-OV-3 cells. However, to date, there has been no research on the functional relationship and molecular mechanism of estrogen-regulating Orai1 during EOC development. In our study, Orai1 had a high expression level in high-grade serous ovarian tumor tissues and SK-OV-3 cells. Estrogen promoted cell proliferation and migration while inhibiting cell apoptosis in SK-OV-3 cells. Orai1 silencing suppressed estrogen-induced cell migration and proliferation. Overexpression of Orai1, however, enhanced the ability of 17β-estradiol (E2) to exert its function. Estrogen induced rapid calcium influx in SK-OV-3 cells. Knockdown of Orai1 in SK-OV-3 cells blocked E2-induced stored-operated Ca2+ influx. The messenger RNA expression of caspase 3, matrix metallopeptidase 1, and cyclin-dependent kinase 6 were regulated via Orai1 under E2 treatment. Our results suggest that estrogen, by regulating Orai1, induced calcium influx to determine cell fate.
Collapse
Affiliation(s)
- Xiaoyu Lv
- Plastic Surgery Institute, Weifang Medical University, Weifang, Shandong, China
| | - Chunlei Miao
- Plastic Surgery Institute, Weifang Medical University, Weifang, Shandong, China
| | - Mengyan Liu
- Plastic Surgery Institute, Weifang Medical University, Weifang, Shandong, China
| | - Xinbo Wang
- Department of Gynecology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Lin Wang
- School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Di Wang
- Plastic Surgery Institute, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
9
|
Gov E. Co-expressed functional module-related genes in ovarian cancer stem cells represent novel prognostic biomarkers in ovarian cancer. Syst Biol Reprod Med 2020; 66:255-266. [DOI: 10.1080/19396368.2020.1759730] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Esra Gov
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| |
Collapse
|
10
|
Xue Y, Bhushan B, Mars WM, Bowen W, Tao J, Orr A, Stoops J, Yu Y, Luo J, Duncan AW, Michalopoulos GK. Phosphorylated Ezrin (Thr567) Regulates Hippo Pathway and Yes-Associated Protein (Yap) in Liver. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1427-1437. [PMID: 32289287 PMCID: PMC10069283 DOI: 10.1016/j.ajpath.2020.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/20/2020] [Accepted: 03/26/2020] [Indexed: 12/18/2022]
Abstract
The activation of CD81 [the portal of entry of hepatitis C virus (HCV)] by agonistic antibody results in phosphorylation of Ezrin via Syk kinase and is associated with inactivation of the Hippo pathway and increase in yes-associated protein (Yap1). The opposite occurs when glypican-3 or E2 protein of HCV binds to CD81. Hepatocyte-specific glypican-3 transgenic mice have decreased levels of phosphorylated (p)-Ezrin (Thr567) and Yap, increased Hippo activity, and suppressed liver regeneration. The role of Ezrin in these processes has been speculated, but not proved. We show that Ezrin has a direct role in the regulation of Hippo pathway and Yap. Forced expression of plasmids expressing mutant Ezrin (T567D) that mimics p-Ezrin (Thr567) suppressed Hippo activity and activated Yap signaling in hepatocytes in vivo and enhanced activation of pathways of β-catenin and leucine rich repeat containing G protein-coupled receptor 4 (LGR4) and LGR5 receptors. Hepatoma cell lines JM1 and JM2 have decreased CD81 expression and Hippo activity and up-regulated p-Ezrin (T567). NSC668394, a p-Ezrin (Thr567) antagonist, significantly decreased hepatoma cell proliferation. We additionally show that p-Ezrin (T567) is controlled by epidermal growth factor receptor and MET. Ezrin phosphorylation, mediated by CD81-associated Syk kinase, is directly involved in regulation of Hippo pathway, Yap levels, and growth of normal and neoplastic hepatocytes. The finding has mechanistic and potentially therapeutic applications in hepatocyte growth biology, hepatocellular carcinoma, and HCV pathogenesis.
Collapse
Affiliation(s)
- Yuhua Xue
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bharat Bhushan
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wendy M Mars
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William Bowen
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anne Orr
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Stoops
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yanping Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jianhua Luo
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew W Duncan
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
11
|
Abstract
Comprehensive theory explaining the relationship between estrogen (E2) and ezrin in metastasis of thyroid cancer remains non-elicited. In vitro results revealed that E2 could stimulate the expression and phosphorylation of ezrin in a time and dose dependent manner. Our data clearly showed that E2 enhanced the migration and invasion of cells, which was reversed by the transfection of cells with ezrin specific siRNA. Further, we observed that Phosphoinositide 3-kinase (PI3K) ROCK-2 are among the kinases responsible for E2 induced phosphorylation of ezrin. Clinical validation of ezrin/phospho-ezrin revealed that phospho-ezrin was intensely expressed in follicular thyroid carcinoma (FTC) and follicular variant of papillary thyroid carcinoma (FVPTC), while it was completely absent in follicular adenoma (FA) lesions in which the differentiation of the follicular neoplasms remains subtle. When histology of different carcinomas is correlated with benign FA with respect to phospho-ezrin, we observed that the marker was highly significant (p = 0.0001). 100% sensitivity, specificity and diagnostic accuracy of the above marker in the histological association of FTC, FVPTC with FA, enables us to suggest phospho-ezrin as a diagnostic marker to differentiate the follicular neoplasms. These data are the first to suggest the dynamic regulation of ezrin phosphorylation during metastasis in FTC.
Collapse
|
12
|
Thomsen LH, Schnack TH, Buchardi K, Hummelshoj L, Missmer SA, Forman A, Blaakaer J. Risk factors of epithelial ovarian carcinomas among women with endometriosis: a systematic review. Acta Obstet Gynecol Scand 2016; 96:761-778. [DOI: 10.1111/aogs.13010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/23/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Line H. Thomsen
- Department of Gynecology and Obstetrics; Aarhus University Hospital; Aarhus Denmark
| | - Tine H. Schnack
- Gynecologic and Obstetric Department; Rigshospitalet University Hospital of Copenhagen; Copenhagen Denmark
| | | | | | - Stacey A. Missmer
- Department of Obstetrics; Gynecology and Reproductive Biology College of Human Medicine; Michigan State University; Boston MA USA
- Department of Epidemiology; Harvard T.H. Chan School of Public Health; Boston MA USA
| | - Axel Forman
- Department of Gynecology and Obstetrics; Aarhus University Hospital; Aarhus Denmark
| | - Jan Blaakaer
- Department of Gynecology and Obstetrics; Aarhus University Hospital; Aarhus Denmark
| |
Collapse
|
13
|
Fadiel A, Choi SD, Park B, Kim TH, Buldo-Licciardi J, Ahmadi M, Arslan A, Mittal K, Naftolin F. Expression of Ezrin and Estrogen Receptors During Cervical Carcinogenesis. Reprod Sci 2016; 24:706-712. [PMID: 27688241 DOI: 10.1177/1933719116667222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
RATIONALE Development of cervical squamous carcinoma (CXCA) is accompanied by changes in estrogen receptors (ERs, ERα and ERβ) and ezrin expression; however, reports have been conflicting. Using histologically documented staging of cervical biopsies, we determined ezrin and ER relationships during CXCA development. METHODS Immunoreactive (ir) ezrin, ir-ERα, and ir-ERβ were studied in normal epithelium, carcinoma in situ/cervical intraepithelial neoplasia (CIN) 1 to 3, and local invasion or metastatic CXCA. Results were compared using H scoring. Cultures of Caski metastatic CXCA cells were treated with estradiol and/or tamoxifen and studied for ER-driven ir-ezrin and the morphologic response. RESULTS Koilocytosis was present and indicated viral presence. The ezrin H score increased from CIN1 to CIN3, reaching significant differences from normal by CIN3 ( P = .004) and 2× normal in metastatic CXCA. Estrogen receptor α and ERβ H scores fell, reaching significance by CIN3 (ERα, P = .0001; ERβ, P = .024). During estradiol treatment, ezrin in Caski cells increased and localized to the periphery, in ruffles and processes. The selective ER modulator tamoxifen blocked the estradiol-induced changes. CONCLUSIONS During cervical carcinogenesis, the usual relationship between estrogen and ezrin induction is abridged. This is consistent with the effects of human papilloma virus viral proteins such as E6 and E7 that upregulate SIX1, a protein that induces ezrin. Cervical carcinogenesis is progressive but arrests at the preinvasive stage for varying lengths of time. These studies suggest that changes in ezrin may be associated with the development of the invasive phenotype and penetration of the basement membrane. They also raise the possibility that inhibiting ezrin expression could be a target for the prevention of invasive CXCA.
Collapse
Affiliation(s)
- Ahmed Fadiel
- 1 Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY, USA
| | - Seung Do Choi
- 1 Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY, USA.,2 Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Cheonan, Republic of Korea
| | - Bora Park
- 3 Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Seoul, Republic of Korea
| | - Tae-Hee Kim
- 4 Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Julia Buldo-Licciardi
- 1 Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY, USA
| | - Mitra Ahmadi
- 1 Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY, USA
| | - Alan Arslan
- 1 Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY, USA
| | - Khushbakhat Mittal
- 1 Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY, USA
| | - Frederick Naftolin
- 1 Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY, USA
| |
Collapse
|
14
|
Naftolin F, Garcia-Segura LM, Horvath TL, Zsarnovszky A, Demir N, Fadiel A, Leranth C, Vondracek-Klepper S, Lewis C, Chang A, Parducz A. Estrogen-Induced Hypothalamic Synaptic Plasticity and Pituitary Sensitization in the Control of the Estrogen-Induced Gonadotrophin Surge. Reprod Sci 2016; 14:101-16. [PMID: 17636222 DOI: 10.1177/1933719107301059] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Proper gonadal function requires coordinated (feedback) interactions between the gonads, adenohypophysis, and brain: the gonads elaborate sex steroids (progestins, androgens, and estrogens) and proteins (inhibin-activin family) during gamete development. In both sexes, the brain-pituitary gonadotrophin-regulating interaction is coordinated by estradiol through its opposing actions on pituitary gonadotrophs (sensitization of the response to gonadotrophin-releasing hormone [GnRH]) versus hypothalamic neurons (inhibition of GnRH secretion). This dynamic tension between the gonadotrophs and the GnRH cells in the brain regulates the circulating gonadotrophins and is termed reciprocal/negative feedback. In females, reciprocal/negative feedback dominates approximately 90% of the ovarian cycle. In a spectacular exception, the dynamic tension is broken during the surge of circulating estrogen that marks follicle and oocyte(s) maturation. The cause is an estradiol-induced disinhibition of the GnRH neurons that releases GnRH secretion to the highly sensitized pituitary gonadotrophs that in turn release the gonadotrophin surge (the estrogen-induced gonadotrophin surge [EIGS], also known as positive feedback). Studies during the past 4 decades have shown this disinhibition to result from estrogen-induced synaptic plasticity (EISP), including a reversible approximately 50% loss in arcuate nucleus synapses. The disinhibited GnRH secretion occurs during maximal gonadotroph sensitization and results in the EIGS. Specific immunoneutralization of estradiol blocks the EISP and EIGS. The EISP is accompanied by increases in insulinlike growth factor 1, polysialylated neural cell adhesion molecule, and ezrin, 3 proteins that the authors believe are the links between estrogen-induced astroglial extension and the EISP that releases GnRH secretion at the moment of maximal sensitization of the pituitary gonadotrophs. The result is the paradoxical surge of gonadotrophins at the peak of ovarian estrogen secretion and the triggering of ovulation. This enhanced understanding of the mechanics of gonadotrophin control clarifies elements of the involved feedback loops and opens the way to a better understanding of the neurobiology of reproduction.
Collapse
Affiliation(s)
- Frederick Naftolin
- Reproductive Neuroscience Unit, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Hoffmann M, Fiedor E, Ptak A. 17β-Estradiol Reverses Leptin-Inducing Ovarian Cancer Cell Migration by the PI3K/Akt Signaling Pathway. Reprod Sci 2016; 23:1600-1608. [PMID: 27255147 DOI: 10.1177/1933719116648214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Accumulating evidence suggests that leptin is expressed at higher levels in obese women and stimulates cell migration in epithelial cancers. However, the biology of ovarian cancer is different from others, mainly due to the production of estrogens because of the involvement of ovarian tissue, which is the main source of estrogens; as a result, the levels are at least 100- to 1000-fold higher than normal circulating levels. Thus, ovarian cancer tissues are exposed to 17β-estradiol, which promotes ovarian cancer cell migration and may modulate the effect of other hormones. Therefore, this study investigated the effects of 17β-estradiol (1 nmol/L) with leptin (1-40 ng/mL) at physiological levels, on the migration of OVCAR-3 and SKOV-3 ovarian cancer cells, and the expression levels and activity of metalloproteinases (MMPs) 2 and 9. Here, we found that leptin stimulated ovarian cancer cell line migration, which is mediated via the expression and activity of MMP-9 in the OVCAR-3 but not in the SKOV-3 cells. After the administration of 17β-estradiol and leptin, we observed antagonistic effects of 17β-estradiol on leptin-induced OVCAR-3 cell migration and MMP-9 expression and activity. Moreover, the antagonistic effect of 17β-estradiol on leptin-induced cancer cell migration was reversed by pretreatment of the cells with the phosphatidylinositol 3-kinase (PI3K) pathway inhibitor. Taken together, our results, for the first time, show that in ovarian cancer cells ObR+/ER+, 17β-estradiol has an antagonistic effect on leptin-induced cell migration as well as MMP-9 expression and activity, which is mediated by the PI3K pathway.
Collapse
Affiliation(s)
- Marta Hoffmann
- Department of Physiology and Toxicology of Reproduction, Chair of Animal Physiology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Elżbieta Fiedor
- Department of Physiology and Toxicology of Reproduction, Chair of Animal Physiology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| | - Anna Ptak
- Department of Physiology and Toxicology of Reproduction, Chair of Animal Physiology, Institute of Zoology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
16
|
Ray JA, Kushnir MM, Rockwood AL, Meikle AW. Direct Measurement of Free Estradiol in Human Serum and Plasma by Equilibrium Dialysis-Liquid Chromatography-Tandem Mass Spectrometry. Methods Mol Biol 2016; 1378:99-108. [PMID: 26602122 DOI: 10.1007/978-1-4939-3182-8_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
We describe a direct method of measurement of free estradiol using equilibrium dialysis followed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Serum aliquots and internal standards are extracted by liquid-liquid extraction using methyl-tert-butyl ether (MTBE) followed by derivatization with dansyl chloride. An API 5500 mass spectrometer operated in positive electrospray mode is used for detection.
Collapse
Affiliation(s)
- Julie A Ray
- ARUP Institute for Clinical and Experimental Pathology, 500 Chipeta Way, Salt Lake City, UT, 84108, USA.
| | - Mark M Kushnir
- ARUP Institute for Clinical and Experimental Pathology, 500 Chipeta Way, Salt Lake City, UT, 84108, USA.
| | - Alan L Rockwood
- ARUP Institute for Clinical and Experimental Pathology, 500 Chipeta Way, Salt Lake City, UT, 84108, USA.
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA.
| | - A Wayne Meikle
- ARUP Institute for Clinical and Experimental Pathology, 500 Chipeta Way, Salt Lake City, UT, 84108, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
- Department of Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
17
|
Na W, Park JW, An JH, Jang J. Size-controllable ultrathin carboxylated polypyrrole nanotube transducer for extremely sensitive 17β-estradiol FET-type biosensors. J Mater Chem B 2016; 4:5025-5034. [DOI: 10.1039/c6tb00897f] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Size-controllable aptamer conjugated ultrathin carboxylated polypyrrole nanotubes (A-UCPPyNTs) were successfully fabricated as transducers in 17β-estradiol field-effect transistor (FET)-type biosensors which has extremely high sensitivity (∼1 fM) and unique selectivity.
Collapse
Affiliation(s)
- Wonjoo Na
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| | - Jin Wook Park
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| | - Ji Hyun An
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| | - Jyongsik Jang
- School of Chemical and Biological Engineering
- Seoul National University
- Seoul 151-742
- Korea
| |
Collapse
|
18
|
Li J, Li L, Li Z, Gong G, Chen P, Liu H, Wang J, Liu Y, Wu X. The role of miR-205 in the VEGF-mediated promotion of human ovarian cancer cell invasion. Gynecol Oncol 2015; 137:125-33. [PMID: 25597268 DOI: 10.1016/j.ygyno.2015.01.531] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/07/2015] [Accepted: 01/12/2015] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Our objective was to investigate a miRNA pathway that acts downstream of VEGF-induced invasion of ovarian cancer cells. METHOD We used two paired high and low metastatic serous ovarian cancer cells to demonstrate the role of miR-205 in VEGF-induced invasion of ovarian cancer cells and to investigate the gene targets of miR-205. RESULTS Our previous comparative proteomics studies showed that VEGF decreased the expression of Ezrin and Lamin A/C, and this result was validated in the present study using qPCR and Western blotting. Then we found that VEGF enhanced the invasiveness of and inhibited apoptosis in ovarian cancer cells as assessed by transwell invasion assays and Annexin V-FITC immunostaining, respectively. VEGFR was also expressed in ovarian cancer cells, as assessed by immunocytochemical staining. Furthermore, using the dual-luciferase report assay system, we demonstrated that miR-205 targeted Ezrin and Lamin A/C. MiR-205 was up-regulated in ovarian cancer cells exposed to VEGF, as determined by miRNA microarray analysis and verified by qPCR. MiR-205 promoted the invasion and proliferation of ovarian cancer cells. CONCLUSION Our data reveal a new potential pathway in which VEGF promotes the invasion of ovarian cancer cells, partially via the down-regulation of Ezrin and Lamin A/C caused by increased expression of miR-205.
Collapse
Affiliation(s)
- Juanni Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Pathology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Long Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Pathology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Zexia Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Microarray Core Facility, University of Texas Southwestern Medical Center, 75070, USA
| | - Guanghui Gong
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Pathology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Puxiang Chen
- Department of Gynecology and Obstetrics, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Hailing Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Pathology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Junpu Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Pathology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Ying Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Pathology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Xiaoying Wu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Pathology, School of Basic Medical Science, Central South University, Changsha 410013, China.
| |
Collapse
|
19
|
Lee HH, Kim TH, Park J, Lee A, Park Y, Byun DW, Kim MJ, Lim H. Expression of ezrin in vagina cells of postmenopausal rats after dietary administration of omega-3 Fatty Acid formula. J Menopausal Med 2015; 20:97-103. [PMID: 25580420 PMCID: PMC4286661 DOI: 10.6118/jmm.2014.20.3.97] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/18/2014] [Accepted: 10/20/2014] [Indexed: 11/21/2022] Open
Abstract
Objectives To see the effect of dietary administration of omega 3-fatty acid formula on the vaginal cells of postmenopausal rats. Methods Three-week-old female Wistar/ST rats were raised after one week of adjustment period. The rats were then divided into three groups, for three different kinds of diet; general diet, 1% omega-3 fatty acid diet, and 2% omega-3 fatty acid diet. After eight weeks of having assigned diet, after the oophorectomy, with the same diet previously they had Immunohistochemistry, Immunofluorescence, and Western Blot about ezrin, merlin were done. Results In immunohistochemistry, estrogen injection group revealed thicker and well differentiated features. In Immunofluorescence, Omega-3 fatty acid composition in diet did not effect expression of ezrin and merlin in rat vagina in estrogen injection group, their vaginal epithelium showed full layers (from basal to apical layer). In Western Blot analysis, Omega-3 fatty acid composition in diet did not affect expression of ezrin and merlin in rat vagina estrogen presented significant impact on expression of ezrin and merlin. Conclusion Although omega-3 fatty acid composition changed in diet, vaginal epithelial morphology unchanged. Estrogen did effect on vagina cell, but omega-3 fatty acid did not effect on ezrin and merlin in vagina.
Collapse
Affiliation(s)
- Hae-Hyeog Lee
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Tae-Hee Kim
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Junsik Park
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Arum Lee
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon, Korea. ; Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang University, Asan, Korea
| | - Yongsoon Park
- Department of Food and Nutrition, College of Human Ecology, Hanyang University, Seoul, Korea
| | - Dong Won Byun
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Min Jung Kim
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon, Korea. ; Department of Food and Nutrition, College of Human Ecology, Hanyang University, Seoul, Korea
| | - Heesook Lim
- Department of Nutrition, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| |
Collapse
|
20
|
Du L, Ji W, Zhang Y, Zhang C, Liu G, Wang S. An ultrasensitive detection of 17β-estradiol using a gold nanoparticle-based fluorescence immunoassay. Analyst 2015; 140:2001-7. [DOI: 10.1039/c4an01952k] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A fluorescence immunoassay based on functionalized gold nanoparticle amplification and immunomagnetic separation was constructed for E2detection.
Collapse
Affiliation(s)
- Lingyun Du
- Shandong Provincial Key Laboratory of Chemical Energy Storage and spectrometry (LC/MS/MS) and gas chromatography-mass Novel Cell Technology
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- China
| | - Wei Ji
- Shandong Provincial Key Laboratory of Chemical Energy Storage and spectrometry (LC/MS/MS) and gas chromatography-mass Novel Cell Technology
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- China
| | - Yuanfu Zhang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and spectrometry (LC/MS/MS) and gas chromatography-mass Novel Cell Technology
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- China
| | - Chunyan Zhang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and spectrometry (LC/MS/MS) and gas chromatography-mass Novel Cell Technology
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- China
| | - Guofu Liu
- College of Life Science
- Liaocheng University
- Liaocheng 252059
- China
| | - Shuhao Wang
- Shandong Provincial Key Laboratory of Chemical Energy Storage and spectrometry (LC/MS/MS) and gas chromatography-mass Novel Cell Technology
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- China
| |
Collapse
|
21
|
Ptak A, Hoffmann M, Gruca I, Barć J. Bisphenol A induce ovarian cancer cell migration via the MAPK and PI3K/Akt signalling pathways. Toxicol Lett 2014; 229:357-65. [DOI: 10.1016/j.toxlet.2014.07.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 11/25/2022]
|
22
|
Estrogen combined with progesterone decreases cell proliferation and inhibits the expression of Bcl-2 via microRNA let-7a and miR-34b in ovarian cancer cells. Clin Transl Oncol 2014; 16:898-905. [PMID: 24643702 DOI: 10.1007/s12094-014-1166-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 02/15/2014] [Indexed: 01/19/2023]
Abstract
PURPOSE This study evaluated the effect of estrogen (E(2)), progesterone (P4), and the combination of them (E(2) + P4) on survival rate, apoptosis, and the expressions of Bcl-2, hsa-let-7a and has-miR-34b in primary ovarian cancer cells to provide new clues for the clinical treatments of ovarian cancer. METHODS The primary ovarian cancer cells from 60 cases of clinical ovarian cancer tissues were isolated and then cultured. The survival rate of ovarian cancer cells after the treatment of E(2), P4 and E(2) + P4 was analyzed by MTT assay. Cell apoptosis rate and cell cycle were measured by FACS analysis. Moreover, the relative abundance of Bcl-2 and microRNAs (let-7a, miR-34b) expressions were detected by quantitative real-time PCR (qRT-PCR) and Western blotting. RESULTS Low concentrations of estrogen (10(-10), 10(-8), 10(-6 )mol/L) did not affect the proliferation of ovarian cancer cells. However, the high concentration of estrogen (10(-4 )mol/L) inhibited survival rate of ovarian cancer cells. Progesterone (10(-4 )mol/L) inhibited the proliferation of cancer cells. The combination of estrogen and progesterone significantly inhibited the survival rate of ovarian cancer cells with a time- and dose-dependent manner. High concentration of estrogen combined with progesterone (E(2) + P4) induced apoptosis of ovarian cancer cells. E(2) + P4 promoted the expression of let-7a and miR-34b and reduced the expression of Bcl-2 in ovarian cancer cells. When the expression of let-7a or/and miR-34b was inhibited using miRNA inhibitors, E(2) + P4 treatment did not change the protein level of Bcl-2. CONCLUSION E(2) + P4 significantly inhibited the cell survival, promoted the cell apoptosis, induced the expression of let-7a and miR-34b, and reduced the expression of Bcl-2 in ovarian cancer cells.
Collapse
|
23
|
Antelmi E, Cardone RA, Greco MR, Rubino R, Di Sole F, Martino NA, Casavola V, Carcangiu M, Moro L, Reshkin SJ. ß1 integrin binding phosphorylates ezrin at T567 to activate a lipid raft signalsome driving invadopodia activity and invasion. PLoS One 2013; 8:e75113. [PMID: 24086451 PMCID: PMC3782503 DOI: 10.1371/journal.pone.0075113] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 08/09/2013] [Indexed: 01/11/2023] Open
Abstract
Extracellular matrix (ECM) degradation is a critical process in tumor cell invasion and requires matrix degrading protrusions called invadopodia. The Na+/H+ exchanger (NHE1) has recently been shown to be fundamental in the regulation of invadopodia actin cytoskeleton dynamics and activity. However, the structural link between the invadopodia cytoskeleton and NHE1 is still unknown. A candidate could be ezrin, a linker between the NHE1 and the actin cytoskeleton known to play a pivotal role in invasion and metastasis. However, the mechanistic basis for its role remains unknown. Here, we demonstrate that ezrin phosphorylated at T567 is highly overexpressed in the membrane of human breast tumors and positively associated with invasive growth and HER2 overexpression. Further, in the metastatic cell line, MDA-MB-231, p-ezrin was almost exclusively expressed in invadopodia lipid rafts where it co-localized in a functional complex with NHE1, EGFR, ß1-integrin and phosphorylated-NHERF1. Manipulation by mutation of ezrins T567 phosphorylation state and/or PIP2 binding capacity or of NHE1s binding to ezrin or PIP2 demonstrated that p-ezrin expression and binding to PIP2 are required for invadopodia-mediated ECM degradation and invasion and identified NHE1 as the membrane protein that p-ezrin regulates to induce invadopodia formation and activity.
Collapse
Affiliation(s)
- Ester Antelmi
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
- Department of Pathology, Anatomic Pathology A Unit, Istituto Nazionale Tumori, Milan, Italy
| | - Rosa A. Cardone
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
| | - Maria R. Greco
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
| | - Rosa Rubino
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
| | - Francesca Di Sole
- Department of Medicine, University of Maryland School of Medicine and the Medical Service, Department of Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Nicola A. Martino
- Department of Animal Production, Faculty of Biotechnological Sciences, University of Bari, Bari, Italy
| | - Valeria Casavola
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
| | - MariaLuisa Carcangiu
- Department of Pathology, Anatomic Pathology A Unit, Istituto Nazionale Tumori, Milan, Italy
| | - Loredana Moro
- Institute of Biomembranes and Bioenergetics (IBBE), CNR, Bari, Italy
| | - Stephan J. Reshkin
- Department of Bioscience, Biotechnology and Biopharmacologics, University of Bari, Bari, Italy
- * E-mail:
| |
Collapse
|
24
|
Karabudak AA, Hafner J, Shetty V, Chen S, Secord AA, Morse MA, Philip R. Autoantibody biomarkers identified by proteomics methods distinguish ovarian cancer from non-ovarian cancer with various CA-125 levels. J Cancer Res Clin Oncol 2013; 139:1757-70. [PMID: 23999876 DOI: 10.1007/s00432-013-1501-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 08/21/2013] [Indexed: 02/06/2023]
Abstract
PURPOSE CA-125 has been a valuable marker for detecting ovarian cancer, however, it is not sensitive enough to detect early-stage disease and not specific to ovarian cancer. The purpose of our study was to identify autoantibody markers that are specific to ovarian cancer regardless of CA-125 levels. METHODS Top-down and iTRAQ quantitative proteomics methods were used to identify high-frequency autoantibodies in ovarian cancer. Protein microarrays comprising the recombinant autoantigens were screened using serum samples from various stages of ovarian cancer with diverse levels of CA-125 as well as benign and healthy controls. ROC curve and dot blot analyses were performed to validate the sensitivity and specificity of the autoantibody markers. RESULTS The proteomics methodologies identified more than 60 potential high-frequency autoantibodies in ovarian cancer. Individual serum samples from ovarian cancer stages I-IV compared to control samples that were screened on a microarray containing native recombinant autoantigens revealed a panel of stage I high-frequency autoantibodies. Preliminary ROC curve and dot blot analyses performed with the ovarian cancer samples showed higher specificity and sensitivity as compared to CA-125. Three autoantibody markers exhibited higher specificity in various stages of ovarian cancer with low and normal CA-125 levels. CONCLUSIONS Proteomics technologies are suitable for the identification of protein biomarkers and also the identification of autoantibody biomarkers when combined with protein microarray screening. Using native recombinant autoantigen arrays to screen autoantibody markers, it is possible to identify markers with higher sensitivity and specificity than CA-125 that are relevant to early detection of ovarian cancer.
Collapse
|
25
|
Li HH, Zhao YJ, Li Y, Dai CF, Jobe SO, Yang XS, Li XF, Patankar MS, Magness RR, Zheng J. Estradiol 17β and its metabolites stimulate cell proliferation and antagonize ascorbic acid-suppressed cell proliferation in human ovarian cancer cells. Reprod Sci 2013; 21:102-11. [PMID: 23757313 DOI: 10.1177/1933719113492211] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Estradiol 17β (E2β) and ascorbic acid (AA) have been implicated in cancer progression. However, little is known about the actions of biologically active metabolites of E2β, 2-hydroxyestradiol (2OHE2), 4-hydroxyestradiol (4OHE2), 2-methoxyestradiol (2ME2), and 4-methoxyestradiol (4ME2) synthesized sequentially by cytochrome P450, family 1, subfamily A (CYP1A1) and B (CYP1B1), polypeptide 1, and catechol-O-methyltransferase (COMT) on ovarian cancer. Herein, we examined the expression of CYP1A1, CYP1B1, COMT, and estrogen receptor α (ERα) and β (ERβ) in human ovarian surface epithelial (IOSE-385) and cancer cell lines (OVCAR-3, SKOV-3, and OVCA-432). We also investigated the roles of E2β, 2OHE2, 4OHE2, 2ME2, and 4ME2 in cell proliferation, and their interactive effects with AA on ovarian cells. We found the expression of CYP1A1, CYP1B1, COMT, ERα, and ERβ in most cell lines tested. Treating cells with physiological concentrations of E2β and its metabolites promoted (13%-42% of the control) IOSE-385 and OVCAR-3 proliferation. The ER blockade inhibited IOSE-385 (∼76%) and OVCAR-3 (∼87%) proliferative response to E2β but not to its metabolites. The ERα blockade inhibited (∼85%) E2β-stimulated OVCAR-3 proliferation, whereas ERβ blockade attenuated (∼83%) E2β-stimulated IOSE-385 proliferation. The AA at ≥250 μmol/L completely inhibited serum-stimulated cell proliferation in all cell lines tested; however, such inhibition in IOSE-385, OVCAR-3, and OVCA-432 was partially (∼10%-20%) countered by E2β and its metabolites. Thus, our findings indicate that E2β and its metabolites promote cell proliferation and antagonize the AA-suppressed cell proliferation in a subset of ovarian cancer cells, suggesting that blocking the actions of E2β and its metabolites may enhance AA's antiovarian cancer activity.
Collapse
Affiliation(s)
- Hui-Hui Li
- 1Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Shandong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yan Y, Liu H, Wen H, Jiang X, Cao X, Zhang G, Liu G. The novel estrogen receptor GPER regulates the migration and invasion of ovarian cancer cells. Mol Cell Biochem 2013; 378:1-7. [PMID: 23580092 DOI: 10.1007/s11010-013-1579-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 01/30/2013] [Indexed: 12/17/2022]
Abstract
G protein-coupled estrogen receptor (GPER) was identified as a new member of the estrogen receptor family in recent years. It has become apparent that GPER mediates the non-genomic signaling of 17β-estradiol (E2) in a variety of estrogen-related cancers. Our previous study has found that GPER was overexpressed in human epithelial ovarian cancer and was positively correlated with the expression of matrix metalloproteinase 9 (MMP-9), which suggested GPER might promote the metastasis of ovarian cancer. However, the mechanisms underlying GPER-dependent metastasis of ovarian cancer are still not clear. In the present study, estrogen receptor α (ERα)-negative/GPER-positive OVCAR5 ovarian cancer cell line was used to investigate the role of GPER in the migration and invasion of ovarian cancer. Wound healing assay and transwell matrigel invasion assay were performed to determine the potentials of cell migration and invasion, respectively. The production and activity of MMP-9 in OVCAR5 cells were examined by Western blot and gelatin zymography analysis. The results showed that E2 and selective GPER agonist G-1 increased cell motility and invasiveness, and upregulated the production and proteolytic activity of MMP-9 in OVCAR5 cells. Small interfering RNA (siRNA) targeting GPER and G protein inhibitor pertussin toxin (PTX) inhibited the migration and invasion of OVCAR5 cells, and also reduced the expression and activity of MMP-9. Our data suggested that GPER promoted the migration and invasion of ovarian cancer cells by increasing the expression and activity of MMP-9. GPER might play an important role in the progression of ovarian cancer.
Collapse
Affiliation(s)
- Yan Yan
- Laboratory of Reproductive Endocrinology, Department of Physiology, Harbin Medical University, Nangang District, Harbin, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Schüler S, Ponnath M, Engel J, Ortmann O. Ovarian epithelial tumors and reproductive factors: a systematic review. Arch Gynecol Obstet 2013; 287:1187-204. [PMID: 23503972 DOI: 10.1007/s00404-013-2784-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 02/28/2013] [Indexed: 12/11/2022]
Abstract
PURPOSE The aim of this systematic review is to summarize the current knowledge about the etiology and pathogenesis of borderline tumors ovarian cancer with special emphasis on the role of endocrine treatments and reproductive factors to establish a foundation for future studies. METHODS We performed a systematic review on the relation between ovarian epithelial tumors (OET) and reproductive factors using the keywords: ovarian cancer, ovarian tumor, ovarian borderline tumor, age at menarche, age at menopause, parity, infertility, PCO syndrome, oral contraception, menopausal hormone therapy, fertility treatment. Totally, 3,290 abstracts were scanned for their relevance in this publication and 127 were finally included. RESULTS The incidence of ovarian epithelial cancer and ovarian borderline tumors is influenced by certain reproductive factors. The strongest protective effects are conferred by parity and use of oral contraceptive pills. Recent molecular biologic and histopathologic studies prove that OET represent a diverse group of tumors, each histologic type with a different genetic background. This is at least partly reflected in epidemiologic and clinical studies showing different risk modulating effects of reproductive factors and endocrine therapies on OET. CONCLUSIONS The etiology and pathogenesis of ovarian cancer are still not fully understood. None of the so far proposed hypothesis on the development of OET can fully account for the epidemiologic and clinical findings in the context of reproductive factors and OET development. Further research approaches are warranted and need to put more weight on the clinical and genetical diversity of OET to yield a more detailed insight into their pathogenesis.
Collapse
Affiliation(s)
- Susanne Schüler
- Department of Obstetrics and Gynecology, University of Regensburg, Caritas-Hospital St. Josef, Landshuter Straße 65, 93053 Regensburg, Germany.
| | | | | | | |
Collapse
|
28
|
Fadiel A, Chen Z, Ulukus E, Ohtani K, Hatami M, Naftolin F. Ezrin Overexpression by Transformed Human Ovarian Surface Epithelial Cells, Ovarian Cleft Cells, and Serous Ovarian Adenocarcinoma Cells. Reprod Sci 2012; 19:797-805. [DOI: 10.1177/1933719111433884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Ahmed Fadiel
- Department of Obstetrics and Gynecology, New York University, New York City, NY, USA
- Departments of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT, USA
| | - Zhaocong Chen
- Departments of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT, USA
- Current Address: Department of Molecular Biology, Tongji University Medical School, Wuhan, China
| | - Emine Ulukus
- Pathology, Yale University School of Medicine, New Haven, CT, USA
- Current Address: Department of Pathology, Eylul University, School of Medicine, Inciralti, Izmir, 35340, Turkey
| | - Kaori Ohtani
- Departments of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT, USA
- Current Address: Department of Obstetrics and Gynecology, Nihon University School of Medicine, Oyaguchi-kamimachi, Itabashi, Tokyo, Japan
| | - Mehrangiz Hatami
- Department of Obstetrics and Gynecology, New York University, New York City, NY, USA
| | - Frederick Naftolin
- Department of Obstetrics and Gynecology, New York University, New York City, NY, USA
- Departments of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
29
|
Arslan AA, Silvera D, Arju R, Giashuddin S, Belitskaya-Levy I, Formenti SC, Schneider RJ. Atypical ezrin localization as a marker of locally advanced breast cancer. Breast Cancer Res Treat 2012; 134:981-8. [PMID: 22415480 DOI: 10.1007/s10549-012-2017-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 03/02/2012] [Indexed: 11/24/2022]
Abstract
Locally advanced breast cancer (LABC) was initially characterized as a large primary tumor (≥5 cm), associated with or without skin or chest-wall involvement, fixed axillary lymph nodes, or disease spread to the ipsilateral internal mammary or supraclavicular nodes. Since 2002, LABC has been reclassified to include smaller stage IIB tumors (2 to <5 cm) with lymph node involvement, or stages IIIA-IIIB (≥5 cm) with or without nodal involvement. Despite the rather common presentation of LABC, it remains a poorly understood and highly variable clinical presentation of breast cancer that is a challenge to treatment. Here, we characterized a panel of breast tumors of known stage, grade, and key clinical-pathological parameters for the expression of the protein ezrin, which is involved in promoting signaling of the PI3K-Akt-mTOR pathway in response to extracellular and tumor micro-environmental signals, and is involved in breast cancer invasion and metastasis. We show that ezrin, which resides primarily in the apical membrane in normal breast epithelium, relocalizes primarily to the cytoplasm in >80 % of traditional (T3) invasive ductal LABC tumors (≥5 cm). Cytoplasmic ezrin is very strongly associated with a single characteristic in breast cancer-large tumor size. In contrast, in large non-malignant fibroadenomas, ezrin staining was similar to that of normal breast epithelium. Small (T1, 1 cm) invasive ductal carcinomas displayed largely apical membrane and perinuclear ezrin localization with weak cytoplasmic staining. Cytoplasmic ezrin localization was also associated with positive lymph node status, but no other clinical-pathological features, including hormone receptor status, histological or nuclear grade of tumor cell. The cytoplasmic relocalization of ezrin may therefore represent a novel marker for large malignant tumor size, reflecting the unique biology of LABC.
Collapse
Affiliation(s)
- Alan A Arslan
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Mak H, Naba A, Varma S, Schick C, Day A, SenGupta SK, Arpin M, Elliott BE. Ezrin phosphorylation on tyrosine 477 regulates invasion and metastasis of breast cancer cells. BMC Cancer 2012; 12:82. [PMID: 22397367 PMCID: PMC3372425 DOI: 10.1186/1471-2407-12-82] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 03/07/2012] [Indexed: 11/11/2022] Open
Abstract
Background The membrane cytoskeletal crosslinker, ezrin, a member of the ERM family of proteins, is frequently over-expressed in human breast cancers, and is required for motility and invasion of epithelial cells. Our group previously showed that ezrin acts co-operatively with the non-receptor tyrosine kinase, Src, in deregulation of cell-cell contacts and scattering of epithelial cells. In particular, ezrin phosphorylation on Y477 by Src is specific to ezrin within the ERM family, and is required for HGF-induced scattering of epithelial cells. We therefore sought to examine the role of Y477 phosphorylation in ezrin on tumor progression. Methods Using a highly metastatic mouse mammary carcinoma cell line (AC2M2), we tested the effect of over-expressing a non-phosphorylatable form of ezrin (Y477F) on invasive colony growth in 3-dimensional Matrigel cultures, and on local invasion and metastasis in an orthotopic engraftment model. Results AC2M2 cells over-expressing Y477F ezrin exhibited delayed migration in vitro, and cohesive round colonies in 3-dimensional Matrigel cultures, compared to control cells that formed invasive colonies with branching chains of cells and numerous actin-rich protrusions. Moreover, over-expression of Y477F ezrin inhibits local tumor invasion in vivo. Whereas orthotopically injected wild type AC2M2 tumor cells were found to infiltrate into the abdominal wall and visceral organs within three weeks, tumors expressing Y477F ezrin remained circumscribed, with little invasion into the surrounding stroma and abdominal wall. Additionally, Y477F ezrin reduces the number of lung metastatic lesions. Conclusions Our study implicates a role of Y477 ezrin, which is phosphorylated by Src, in regulating local invasion and metastasis of breast carcinoma cells, and provides a clinically relevant model for assessing the Src/ezrin pathway as a potential prognostic/predictive marker or treatment target for invasive human breast cancer.
Collapse
Affiliation(s)
- Hannah Mak
- Division of Cancer Biology and Genetics, Cancer Research Institute, Queen's University, Kingston, ON, K7L 3N6, Canada
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Direct measurement of free estradiol in human serum by equilibrium dialysis-liquid chromatography-tandem mass spectrometry and reference intervals of free estradiol in women. Clin Chim Acta 2012; 413:1008-14. [PMID: 22421268 DOI: 10.1016/j.cca.2012.02.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 02/27/2012] [Accepted: 02/29/2012] [Indexed: 11/21/2022]
Abstract
BACKGROUND Measurement of free estradiol offers a better representation of the bioactive fraction of the hormone. We describe a direct equilibrium dialysis-liquid chromatography-tandem mass spectrometry (ED-LC-MS/MS) method for serum free estradiol. METHODS Two hundred fifty microliter aliquots of serum were dialyzed for 22h followed by liquid-liquid extraction and derivatization with dansyl chloride. Free estradiol was measured using LC-MS/MS with an AB SCIEX 5500 mass spectrometer in positive ion and multiple reaction monitoring (MRM) mode. RESULTS The limits of detection and quantification for free estradiol were 0.25 and 0.5pg/ml (0.9 and 1.8pmol/l) respectively. Total imprecision was less than 10%. Results of method comparison showed 3 times overestimation using indirect methods of measurement. Reference intervals in pre-menopausal women in follicular, mid-cycle, and luteal phases of cycle were <2.4, <3.1 and <2.6pg/ml (8.8, 11.4, 9.5pmol/l) respectively; in post menopausal women the concentrations were ≤0.5pg/ml (1.8pmol/l). CONCLUSIONS ED-LC-MS/MS is a direct method for accurately measuring free estradiol, independent of total estradiol or sex hormone binding globulin concentrations. Imprecision and sensitivity of the method are adequate for clinical diagnostic applications. The degree of variation observed in the method comparison reinforces the relevance of method specific reference ranges.
Collapse
|
32
|
Tan O, Ornek T, Fadiel A, Carrick KS, Arici A, Doody K, Carr BR, Naftolin F. Expression and activation of the membrane-cytoskeleton protein ezrin during the normal endometrial cycle. Fertil Steril 2012; 97:192-9.e2. [PMID: 22192140 DOI: 10.1016/j.fertnstert.2011.10.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 10/24/2011] [Accepted: 10/31/2011] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To examine total ezrin expression (ezrin and phospho-ezrin) through the normal endometrial cycle and to correlate ezrin activation and localization with cytologic changes. DESIGN Experimental laboratory study. SETTING University medical centers. PATIENT(S) Reproductive-age women. INTERVENTION(S) A total of 36 samples of normal early, mid-, and late proliferative- and secretory-phase endometrium were studied for immunoreactive total ezrin (ir-T-ezrin) and phospho-ezrin (ir-p-ezrin) expression by histology, immunohistochemistry, and Western blotting. MAIN OUTCOME MEASURE(S) Total ezrin and phospho-ezrin expressions through the normal endometrial cycle. RESULT(S) Throughout the cycle ir-T-ezrin is present in the epithelium. The intensity and localization of both ir-ezrin and ir-p-ezrin vary greatly throughout the cycle. The main findings include the following: lateral localization of ir-ezrin/ir-p-ezrin in association with membrane specializations; dense staining around secretory vacuoles (secretory phase); dense staining of the apical surfaces, including microvilli and pinopodes of epithelial cells, especially during the mid- to late secretory phases; and the presence of ezrin in the glandular secretions. Immunoreactive total ezrin and ir-p-ezrin were not expressed by stromal fibroblasts. CONCLUSION(S) Ezrin is a prominent protein in the cycling endometrium. The most striking findings were the gravitation of ir-ezrin/ir-p-ezrin to the periphery of secretory vacuoles, localization on apical surfaces of the luminal epithelium, dense ezrin staining in secretory-phase epithelial cell plumes, and the presence of ir-ezrin/ir-p-ezrin in secretory-phase luminal secretions. These findings may have functional implications, especially for implantation biology.
Collapse
Affiliation(s)
- Orkun Tan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Sanchez AM, Flamini MI, Polak K, Palla G, Spina S, Mannella P, Genazzani AD, Simoncini T. Actin cytoskeleton remodelling by sex steroids in neurones. J Neuroendocrinol 2012; 24:195-201. [PMID: 22103470 DOI: 10.1111/j.1365-2826.2011.02258.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cell morphology and its interaction with the extracellular environment are integrated processes involving a number of intracellular controllers orchestrating cytoskeletal proteins and their interaction with the cell membrane and anchorage proteins. Sex steroids are effective regulators of cell morphology and tissue organisation, and recent evidence indicates that this is obtained through the regulation of the actin cytoskeleton. Intriguingly, many of these regulatory actions related to cell morphology are achieved through the rapid, nonclassical signalling of sex steroid receptors to kinase cascades, independently from nuclear alteration of gene expression or protein synthesis. The identification of the mechanistic basis for these rapid actions on cell cytoskeleton has special relevance for the characterisation of the effects of sex steroids under physiological conditions, such as for the development of neurone/neurone interconnections and dendritic spine density. This is considered to be critical for gender-specific differences in brain function and dysfunction. Recent advancements in the characterisation of the molecular basis of the extranuclear signalling of sex steroids help to clarify the role of oestrogen and progesterone in the brain, and may turn out to be of relevance for clinical purposes. This review highlights the regulatory effects of oestrogens and progesterone on actin cytoskeleton and neurone morphology, as well as recent progresses in the characterisation of these mechanisms, providing insights and working hypotheses on possible clinical applications for the modulation of these pathways in the central nervous system.
Collapse
Affiliation(s)
- A M Sanchez
- Institute of Medicine and Experimental Biology of Cuyo, CCT-CONICET Mendoza, National University of Cuyo, Parque General San Martin s/n, Mendoza, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Ren L, Hong SH, Chen QR, Briggs J, Cassavaugh J, Srinivasan S, Lizardo MM, Mendoza A, Xia AY, Avadhani N, Khan J, Khanna C. Dysregulation of ezrin phosphorylation prevents metastasis and alters cellular metabolism in osteosarcoma. Cancer Res 2011; 72:1001-12. [PMID: 22147261 DOI: 10.1158/0008-5472.can-11-0210] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Ezrin links the plasma membrane to the actin cytoskeleton where it plays a pivotal role in the metastatic progression of several human cancers; however, the precise mechanistic basis for its role remains unknown. Here, we define transitions between active (phosphorylated open) and inactive (dephosphorylated closed) forms of Ezrin that occur during metastatic progression in osteosarcoma. In our evaluation of these conformations we expressed C-terminal mutant forms of Ezrin that are open (phosphomimetic T567D) or closed (phosphodeficient T567A) and compared their biologic characteristics to full-length wild-type Ezrin in osteosarcoma cells. Unexpectedly, cells expressing open, active Ezrin could form neither primary orthotopic tumors nor lung metastases. In contrast, cells expressing closed, inactive Ezrin were also deficient in metastasis but were unaffected in their capacity for primary tumor growth. By imaging single metastatic cells in the lung, we found that cells expressing either open or closed Ezrin displayed increased levels of apoptosis early after their arrival in the lung. Gene expression analysis suggested dysregulation of genes that are functionally linked to carbohydrate and amino acid metabolism. In particular, cells expressing closed, inactive Ezrin exhibited reduced lactate production and basal or ATP-dependent oxygen consumption. Collectively, our results suggest that dynamic regulation of Ezrin phosphorylation at amino acid T567 that controls structural transitions of this protein plays a pivotal role in tumor progression and metastasis, possibly in part by altering cellular metabolism.
Collapse
Affiliation(s)
- Ling Ren
- Tumor and Metastasis Biology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zheng S, Huang J, Zhou K, Zhang C, Xiang Q, Tan Z, Wang T, Fu X. 17β-Estradiol enhances breast cancer cell motility and invasion via extra-nuclear activation of actin-binding protein ezrin. PLoS One 2011; 6:e22439. [PMID: 21818323 PMCID: PMC3144228 DOI: 10.1371/journal.pone.0022439] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 06/27/2011] [Indexed: 12/21/2022] Open
Abstract
Estrogen promotes breast cancer metastasis. However, the detailed mechanism remains largely unknown. The actin binding protein ezrin is a key component in tumor metastasis and its over-expression is positively correlated to the poor outcome of breast cancer. In this study, we investigate the effects of 17β-estradiol (E2) on the activation of ezrin and its role in estrogen-dependent breast cancer cell movement. In T47-D breast cancer cells, E2 rapidly enhances ezrin phosphorylation at Thr567 in a time- and concentration-dependent manner. The signalling cascade implicated in this action involves estrogen receptor (ER) interaction with the non-receptor tyrosine kinase c-Src, which activates the phosphatidylinositol-3 kinase/Akt pathway and the small GTPase RhoA/Rho-associated kinase (ROCK-2) complex. E2 enhances the horizontal cell migration and invasion of T47-D breast cancer cells in three-dimensional matrices, which is reversed by transfection of cells with specific ezrin siRNAs. In conclusion, E2 promotes breast cancer cell movement and invasion by the activation of ezrin. These results provide novel insights into the effects of estrogen on breast cancer progression and highlight potential targets to treat endocrine-sensitive breast cancers.
Collapse
Affiliation(s)
- Shuhui Zheng
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jinghe Huang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kewen Zhou
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chengxi Zhang
- Department of Cardiovascular Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiuling Xiang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi Tan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tinghuai Wang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XDF); (THW)
| | - Xiaodong Fu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- * E-mail: (XDF); (THW)
| |
Collapse
|
36
|
Sanchez AM, Simoncini T. Extra-nuclear signaling of ERalpha to the actin cytoskeleton in the central nervous system. Steroids 2010; 75:528-32. [PMID: 20018203 DOI: 10.1016/j.steroids.2009.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 11/30/2009] [Accepted: 12/02/2009] [Indexed: 11/19/2022]
Abstract
Cell morphology is controlled by a complex and redundant array of intracellular signaling pathways devoted to the regulation of the actin cytoskeleton and of its relationship with the cell membrane and the extracellular matrix. Sex steroids are effective regulators of cell morphology and tissue organization, and recent evidence indicates that this is obtained through the regulation of the cytoskeleton. Intriguingly, many of these regulatory actions related to cell morphology are achieved through rapid, non-classical signaling of sex steroid receptors to kinase cascades, independently from nuclear alteration of gene expression or protein synthesis. The identification of the mechanistic basis for these rapid actions on cell cytoskeleton has special relevance for the characterization of the effects of sex steroids in physiological conditions, such as their role in the control of brain cell remodeling. Brain cell morphology is controlled by estrogens that regulate the development of neuron/neuron interconnections and dendritic spine density. This is thought to be critical for gender-specific differences in brain function and dysfunction. The recent advancements in the characterization of the molecular basis of the extra-nuclear signaling of estrogen helps to understand the role of estrogen in the brain, and may in the future turn out to be of relevance for clinical purposes. This review highlights the regulatory effects on the cytoskeleton and cell morphology of estrogens as well as the recent advances in the characterization of these mechanisms, providing insights and working hypotheses on possible clinical applications for the modulation of these pathways in the central nervous system.
Collapse
Affiliation(s)
- Angel Matias Sanchez
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa, Pisa 56100, Italy
| | | |
Collapse
|
37
|
Li ZB, Chen YX, Zhao JY, Lu J. Effects of pharmacological concentrations of estrogens on growth of 3AO human ovarian cancer cells. ACTA ACUST UNITED AC 2009; 33:782-92. [PMID: 16980124 DOI: 10.1016/s0379-4172(06)60111-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2005] [Accepted: 12/15/2005] [Indexed: 10/24/2022]
Abstract
During the past two decades, the knowledge of the molecular mechanism by which estrogens exert various functions in different tissues and organs has evolved rapidly. Recent reports demonstrated that estrogen could decrease the cell growth in several types of cancer cells, including ovarian cancer cells. Though experiments explored the possible mechanism of the inhibitory effect, the exact mechanism is responsible for the effect, which remains unclear. The ovary is the main source of the estrogen, estrogen receptor is expressed in several ovarian cell types, including ovarian surface epithelium, the tissue of origin of approximately 90% of the ovarian cancers. It was of great interest to analyze the effects of 17beta-estradiol (E2) on apoptosis of ovarian cancer cells, and the identification of E2-regulated specific genes involved in epithelial proliferation apoptosis, thus may be a clue for understanding the progression of ovarian cancer and for the design of new target therapies. To elucidate the mechanism involved, effects of pharmacological concentrations of estrogen were studied in human ovarian cancer cell line 3AO cells. Inhibition of cellular growth of 3AO cells was seen with E2 at concentrations higher than 0.1 micromol/L. The estrogen receptor inhibitor ICI 182780 cannot block the inhibitory effect of E2. It was surprising to find that ICI 182780 itself can inhibit the growth of 3AO cells, and had a collaborative effect with E2. The decreased cell growth induced by E2 was shown to be apoptosis as analyzed by flow cytometry. ERbeta was detected in the 3AO ovarian cancer cell line but not ERalpha. The expression of ERbeta was weak, which may partially explain why high but not low dose of E2 needed to induce the apoptosis of 3AO cells. We also observed that membrane impermeable E2, E2-BSA have lost growth inhibitory on 3AO cells, which excluded the membrane effect of E2 as previously reported by many investigators. The p38 kinase inhibitor, SB203580 were partially protected 3AO cells against growth inhibition by E2, while inhibitor of JNK, SP600125 enhanced cell death induced by E2. These results showed that MAPK is implicated in cellular processes involving apoptosis.
Collapse
Affiliation(s)
- Zong-Bin Li
- Department of Pathophysiology, Second Military Medical University, Shanghai 200433, China
| | | | | | | |
Collapse
|
38
|
Armaiz-Pena GN, Mangala LS, Spannuth WA, Lin YG, Jennings NB, Nick AM, Langley RR, Schmandt R, Lutgendorf SK, Cole SW, Sood AK. Estrous cycle modulates ovarian carcinoma growth. Clin Cancer Res 2009; 15:2971-8. [PMID: 19383821 DOI: 10.1158/1078-0432.ccr-08-2525] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The effects of reproductive hormones on ovarian cancer growth are not well understood. Here, we examined the effects of estrous cycle variation and specific reproductive hormones on ovarian cancer growth. EXPERIMENTAL DESIGN We investigated the role of reproductive hormones in ovarian cancer growth using both in vivo and in vitro models of tumor growth. RESULTS In vivo experiments using the HeyA8 and SKOV3ip1 ovarian cancer models showed that tumor cell inoculation during proestrus significantly increased tumor burden (251-273%) compared with injection during the estrus phase. Treatment of ovariectomized mice with 17beta-estradiol resulted in a 404% to 483% increase in tumor growth compared with controls. Progestins had no significant effect, but did block estrogen-stimulated tumor growth. Tumors collected from mice sacrificed during proestrus showed increased levels of vascular endothelial growth factor (VEGF) and microvessel density compared with mice injected during estrus. HeyA8, SKOV3ip1, and mouse endothelial (MOEC) cells expressed estrogen receptor alpha and beta and progesterone receptor at the protein and mRNA levels, whereas 2774 ovarian cancer cells were estrogen receptor-negative. In vitro assays showed that 17beta-estradiol significantly increased ovarian cancer cell adhesion to collagen in estrogen receptor-positive, but not in estrogen receptor-negative cells. Additionally, 17beta-estradiol increased the migratory potential of MOEC cells, which was abrogated by the mitogen-activated protein kinase (MAPK) inhibitor, PD 09859. Treatment with 17beta-estradiol activated MAPK in MOEC cells, but not in HeyA8 or SKOV3ip1 cells. CONCLUSION Our data suggest that estrogen may promote in vivo ovarian cancer growth, both directly and indirectly, by making the tumor microenvironment more conducive for cancer growth.
Collapse
Affiliation(s)
- Guillermo N Armaiz-Pena
- Department of Gynecologic Oncology and Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Li YF, Hu W, Fu SQ, Li JD, Liu JH, Kavanagh JJ. Aromatase inhibitors in ovarian cancer: is there a role? Int J Gynecol Cancer 2008; 18:600-14. [PMID: 17894799 DOI: 10.1111/j.1525-1438.2007.01075.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Estrogen plays a role in ovarian tumorigenesis. Aromatase is the enzyme required for the synthesis of estrogen via conversion of androgen to estrogen, which is the major source of estrogen in postmenopausal women. Aromatase is present in normal ovaries and other tissues (e.g., fat and muscle) as well as in 33-81% tumor tissues of ovarian cancer. Aromatase inhibitors (AIs) block estrogen synthesis by inhibiting aromatase activity. In patients with recurrent ovarian cancer, single-agent AI therapy has been shown to elicit clinical response rates of up to 35.7% and stable disease rates of 20-42%. Given the limited treatment options for recurrent ovarian cancer and the favorable safety profile and convenient use, AI is a rational option for prolonging platinum-free interval in recurrent ovarian cancer. Further studies are required to determine the efficacy of combination treatment with AIs and biological agents, determine the benefit of AIs for treating special types of ovarian cancer (e.g., endometrioid type), and identify biomarkers for targeted patient selection. This review summarizes the current epidemiologic, preclinical, and clinical data regarding estrogen's role in ovarian cancer, the expression and regulation of aromatase in this disease, the development and characteristics of the three generations of AIs, and the preclinical and clinical studies of AIs in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Y F Li
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77230, USA
| | | | | | | | | | | |
Collapse
|
40
|
Park SH, Cheung LWT, Wong AST, Leung PCK. Estrogen regulates Snail and Slug in the down-regulation of E-cadherin and induces metastatic potential of ovarian cancer cells through estrogen receptor alpha. Mol Endocrinol 2008; 22:2085-98. [PMID: 18550773 DOI: 10.1210/me.2007-0512] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tumorigenesis is a multistep process involving dysregulated cell growth and metastasis. Considerable evidence implicates a mitogenic action of estrogen in early ovarian carcinogenesis. In contrast, its influence in the metastatic cascade of ovarian tumor cells remains obscure. In the present study, we showed that 17beta-estradiol (E2) increased the metastatic potential of human epithelial ovarian cancer cell lines. E2 treatment led to clear morphological changes characteristic of epithelial-mesenchymal transition (EMT) and an enhanced cell migratory propensity. These morphological and functional alterations were associated with changes in the abundance of EMT-related genes. Upon E2 stimulation, expression and promoter activity of the epithelial marker E-cadherin were strikingly suppressed, whereas EMT-associated transcription factors, Snail and Slug, were significantly up-regulated. This up-regulation was attributed to the increase in gene transcription activated by E2. Depletion of endogenous Snail or Slug using small interfering RNA (siRNA) attenuated E2-mediated decrease in E-cadherin. In addition, E2-induced cell migration was also neutralized by the siRNAs, suggesting that both transcription factors are indispensable for the prometastatic actions of E2. More importantly, by using selective estrogen receptor (ER) agonists, forced expression, and siRNA approaches, we identified that E2 triggered the metastatic behaviors exclusively through an ERalpha-dependent pathway. We also showed that ERbeta had an opposing action on ERalpha because the presence of ERbeta completely inhibited the EMT and down-regulation of E-cadherin induced by ERalpha. Collectively, this study provides a compelling argument that estrogen can potentiate tumor progression by EMT induction and highlights the crucial role of ERalpha in ovarian tumorigenesis.
Collapse
Affiliation(s)
- Se-Hyung Park
- Department of Obstetrics and Gynecology, University of British Columbia, 2H-30, 4490 Oak Street, Vancouver, British Columbia, Canada V6H 3V5
| | | | | | | |
Collapse
|
41
|
Giretti MS, Fu XD, De Rosa G, Sarotto I, Baldacci C, Garibaldi S, Mannella P, Biglia N, Sismondi P, Genazzani AR, Simoncini T. Extra-nuclear signalling of estrogen receptor to breast cancer cytoskeletal remodelling, migration and invasion. PLoS One 2008; 3:e2238. [PMID: 18493596 PMCID: PMC2375059 DOI: 10.1371/journal.pone.0002238] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Accepted: 03/31/2008] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Estrogen is an established enhancer of breast cancer development, but less is known on its effect on local progression or metastasis. We studied the effect of estrogen receptor recruitment on actin cytoskeleton remodeling and breast cancer cell movement and invasion. Moreover, we characterized the signaling steps through which these actions are enacted. METHODOLOGY/PRINCIPAL FINDINGS In estrogen receptor (ER) positive T47-D breast cancer cells ER activation with 17beta-estradiol induces rapid and dynamic actin cytoskeleton remodeling with the formation of specialized cell membrane structures like ruffles and pseudopodia. These effects depend on the rapid recruitment of the actin-binding protein moesin. Moesin activation by estradiol depends on the interaction of ER alpha with the G protein G alpha(13), which results in the recruitment of the small GTPase RhoA and in the subsequent activation of its downstream effector Rho-associated kinase-2 (ROCK-2). ROCK-2 is responsible for moesin phosphorylation. The G alpha(13)/RhoA/ROCK/moesin cascade is necessary for the cytoskeletal remodeling and for the enhancement of breast cancer cell horizontal migration and invasion of three-dimensional matrices induced by estrogen. In addition, human samples of normal breast tissue, fibroadenomas and invasive ductal carcinomas show that the expression of wild-type moesin as well as of its active form is deranged in cancers, with increased protein amounts and a loss of association with the cell membrane. CONCLUSIONS/SIGNIFICANCE These results provide an original mechanism through which estrogen can facilitate breast cancer local and distant progression, identifying the extra-nuclear G alpha(13)/RhoA/ROCK/moesin signaling cascade as a target of ER alpha in breast cancer cells. This information helps to understand the effects of estrogen on breast cancer metastasis and may provide new targets for therapeutic interventions.
Collapse
Affiliation(s)
- Maria Silvia Giretti
- Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
| | - Xiao-Dong Fu
- Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
| | - Giovanni De Rosa
- Pathology Unit, University of Turin, Mauriziano “Umberto I” Hospital and Institute for Cancer Research and Treatment of Candiolo, Turin, Italy
| | - Ivana Sarotto
- Pathology Unit, University of Turin, Mauriziano “Umberto I” Hospital and Institute for Cancer Research and Treatment of Candiolo, Turin, Italy
| | - Chiara Baldacci
- Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
| | - Silvia Garibaldi
- Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
| | - Paolo Mannella
- Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
| | - Nicoletta Biglia
- Department of Gynecological Oncology, University of Turin, Mauriziano “Umberto I” Hospital and Institute for Cancer Research and Treatment of Candiolo, Turin, Italy
| | - Piero Sismondi
- Department of Gynecological Oncology, University of Turin, Mauriziano “Umberto I” Hospital and Institute for Cancer Research and Treatment of Candiolo, Turin, Italy
| | | | - Tommaso Simoncini
- Department of Reproductive Medicine and Child Development, University of Pisa, Pisa, Italy
- * E-mail:
| |
Collapse
|
42
|
Fadiel A, Lee HH, Demir N, Richman S, Iwasaki A, Connell K, Naftolin F. Ezrin is a key element in the human vagina. Maturitas 2008; 60:31-41. [PMID: 18486367 DOI: 10.1016/j.maturitas.2008.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Revised: 03/14/2008] [Accepted: 03/17/2008] [Indexed: 12/01/2022]
Abstract
OBJECTIVE The vagina is a complex tubular structure that has reproductive, support and barrier functions. These depend on the cytoarchitecture of the vaginal cells, which is controlled by key proteins. Cytoskeletal proteins determine cell polarity and membrane specializations by integrating the actin cytoskeleton with cell membranes. This integration is the domain of cytoskeletal proteins including the MERM protein family (moesin-ezrin-radixin-Merlin). Nothing is known about the cyto-localization of the MERM's in the vaginal epithelium or how it influences the cytoarchitecture of the vaginal epithelium and stroma. DESIGN Full-thickness human vaginal fornix samples were obtained from 20 normal human specimens obtained at surgery for pelvic relaxation. Light- and electron microscopical immunohistochemistry (IHC) were used to identify and study activation and cellular localization of immuno-reactive-ezrin (ir-ezrin), a prototypical MERM. RESULTS Ir-ezrin was identified in the stratified squamous vaginal epithelium and connective tissue (fibroblasts, blood vessels and leucocytes). "H" scoring indicated that ir-ezrin staining is denser in the vaginal epithelium than in other layers, that the ir-ezrin staining was associated with increased keratinization and with the size of the tight junctions (p<0.01). Both the amounts and localization of ir-ezrin were associated with high levels of estrogen, identified by the menstrual history and keratinization of the superficial vaginal epithelium. The density of stromal ir-ezrin was increased in the presence of dense epithelial keratinization. Immuno-reactive-ezrin staining was most pronounced near the cell membranes of both keratinized and non-keratinized epithelium, indicating that ezrin activation (unfolding and movement to the membrane) had occurred. Ultra-structural examination of the epithelium showed intra-cellular ir-ezrin to be localized to junctional complexes that have been associated with decreased mucosal penetration by microorganisms. Ir-ezrin was widely distributed throughout stromal fibro-muscular cell, vessels and immunocytes. CONCLUSIONS MERM's, represented by ezrin, are widely present in the vaginal wall. This has implications for the strength and resilience of this tubular structure and may be the case in other internal genital tissues. Ezrin's localization and association with cell specializations indicate that in the vagina, as in other tissues, ezrin likely modulates vaginal cell-cell interactions including the changing vaginal cellular interface with the external environment, the regulation of the elasticity of the vagina, and the regulation of microbial and chemical traffic that determine the pH and microbial environment of the vagina. In other work we have shown that ezrin expression is induced by estradiol. The increase of ir-ezrin staining during the appearance of keratinization and maturation of the vaginal cytology indicates that estrogen may regulate vaginal ezrin and thereby the properties of the vaginal wall and epithelium.
Collapse
Affiliation(s)
- A Fadiel
- Department of Obstetrics and Gynecology, New York University, 550 First Avenue, TH528, New York, NY 10016, United States
| | | | | | | | | | | | | |
Collapse
|
43
|
Chen CL, Cheung LWT, Lau MT, Choi JH, Auersperg N, Wang HS, Wong AST, Leung PCK. Differential role of gonadotropin-releasing hormone on human ovarian epithelial cancer cell invasion. Endocrine 2007; 31:311-20. [PMID: 17906381 DOI: 10.1007/s12020-007-0041-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Revised: 06/07/2007] [Accepted: 07/05/2007] [Indexed: 01/03/2023]
Abstract
Ovarian cancer is the most lethal of all gynecological cancers. Most deaths from ovarian cancer are due to widespread intraperitoneal metastases and malignant ascites. However, mechanisms of invasion in ovarian cancer remain poorly understood. In this study, we examined the effects of gonadotropin-releasing hormone (GnRH)-I (the classical mammalian GnRH), GnRH-II (a second form of GnRH), and GnRH receptor on invasion using two human ovarian carcinoma cell lines, OVCAR-3 and SKOV-3. Here we demonstrated that in OVCAR-3, GnRH-I and GnRH-II promoted cell invasion, whereas in SKOV-3, GnRH-I and GnRH-II inhibited cell invasion. Transfection of small interfering RNA to abrogate the gene expression of GnRH receptor reversed GnRH-I and GnRH-II-mediated invasion activities, suggesting that the same receptor, type I GnRH receptor, is essential for the effects of GnRH-I and GnRH-II in both OVCAR-3 and SKOV-3. Treatment of SKOV-3 cells with GnRH-I or GnRH-II resulted in a decrease in matrix metalloproteinase 2 but an increase in tissue inhibitor of metalloproteinase 2 secretions. In addition, we found that GnRH-I and GnRH-II interfered with activation of the phosphatidylinositol-3-kinase/AKT pathway that is well documented to stimulate proteolysis and invasion of ovarian cancer cells. Taken together, these observations suggest that GnRH-I and GnRH-II play key regulatory roles in ovarian tumor cell invasion and extracellular matrix degradation.
Collapse
Affiliation(s)
- Chien-Lin Chen
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taiwan, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Ovarian carcinoma continues to be the leading cause of death due to gynecological malignancy. Epidemiologic studies indicate that steroid hormones play roles in ovarian carcinogenesis. Gonadotropins, estrogen, and androgen may be causative factors, while gonadotropin-releasing hormone and progesterone may be protective factors in ovarian cancer pathogenesis. Experimental studies have shown that hormonal receptors are expressed in ovarian cancer cells and mediate the growth-stimulatory or growth-inhibitory effects of the hormones on these cells. Hormonal therapeutic agents have been evaluated in several clinical trials. Most of these trials were conducted in patients with recurrent or refractory ovarian cancer, with modest efficacy and few side effects. Better understanding of the mechanisms through which hormones affect cell growth may improve the efficacy of hormonal therapy. Molecular markers that can reliably predict major clinical outcomes should be investigated further in well-designed trials.
Collapse
Affiliation(s)
- H Zheng
- Department of Gynecologic Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77230-1439, USA
| | | | | | | | | |
Collapse
|
45
|
Ou K, Kesuma D, Ganesan K, Yu K, Soon SY, Lee SY, Goh XP, Hooi M, Chen W, Jikuya H, Ichikawa T, Kuyama H, Matsuo EI, Nishimura O, Tan P. Quantitative profiling of drug-associated proteomic alterations by combined 2-nitrobenzenesulfenyl chloride (NBS) isotope labeling and 2DE/MS identification. J Proteome Res 2006; 5:2194-206. [PMID: 16944931 DOI: 10.1021/pr060115n] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The identification of drug-responsive biomarkers in complex protein mixtures is an important goal of quantitative proteomics. Here, we describe a novel approach for identifying such drug-induced protein alterations, which combines 2-nitrobenzenesulfenyl chloride (NBS) tryptophan labeling with two-dimensional gel electrophoresis (2DE)/mass spectrometry (MS). Lysates from drug-treated and control samples are labeled with light or heavy NBS moiety and separated on a common 2DE gel, and protein alterations are identified by MS through the differential intensity of paired NBS peptide peaks. Using NBS/2DE/MS, we profiled the proteomic alterations induced by tamoxifen (TAM) in the estrogen receptor (ER) positive MCF-7 breast cancer cell line. Of 88 protein spots that significantly changed upon TAM treatment, 44 spots representing 23 distinct protein species were successfully identified with NBS-paired peptides. Of these 23 TAM-altered proteins, 16 (70%) have not been previously associated with TAM or ER activity. We found the NBS labeling procedure to be both technically and biologically reproducible, and the NBS/2DE/MS alterations exhibited good concordance with conventional 2DE differential protein quantitation, with discrepancies largely due to the comigration of distinct proteins in the regular 2DE gels. To validate the NBS/2DE/MS results, we used immunoblotting to confirm GRP78, CK19, and PA2G4 as bona fide TAM-regulated proteins. Furthermore, we demonstrate that PA2G4 expression can serve as a novel prognostic factor for disease-free survival in two independent breast cancer patient cohorts. To our knowledge, this is the first report describing the proteomic changes in breast cancer cells induced by TAM, the most commonly used selective estrogen receptor modulator (SERM). Our results indicate that NBS/2DE/MS may represent a more reliable approach for cellular protein quantitation than conventional 2DE approaches.
Collapse
Affiliation(s)
- Keli Ou
- Agenica Research/National Cancer Centre/Genome Institute of Singapore, 11 Hospital Drive, Singapore 169610
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Beckner ME, Zhang Z, Agostino NR, Day BW, Pollack IF. Albumin marks pseudopodia of astrocytoma cells responding to hepatocyte growth factor or serum. J Transl Med 2006; 86:1103-14. [PMID: 16969371 DOI: 10.1038/labinvest.3700470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
It is well accepted that dysfunction in the blood brain barrier (BBB) allows permeation of albumin from the bloodstream into astrocytic brain tumors, especially glioblastomas, the most aggressive astrocytomas. In vitro, bovine serum albumin (BSA) aids functional cell assays by maintaining cytokines and growth factors in solution and delivering its cargo of fatty acids. Earlier, we showed that BSA was prominent in lysates prepared from pseudopodia formed by U87 astrocytoma cells. The present studies investigated the association of albumin with pseudopodia formed by U87 and LN229 astrocytoma cells. With hepatocyte growth factor (HGF) stimulation, cell migration was enhanced and BSA, especially its dimerized form, was prominent in pseudopodia compared to unmigrated cells on one-dimensional gels and immunoblots. When lysates were equalized for levels of glyceraldehyde-3-phosphate dehydrogenase, the rise for BSA levels in pseudopodia vs migrated cells was comparable or greater than levels noted for established pseudopodial proteins, beta-actin and ezrin. The increase for dimerized BSA in pseudopodia compared to unmigrated cells was greater than the rise in levels of beta-actin, ezrin, HGF, and phosphorylated Met when pseudopodia were harvested from filters with 1 mum pores using either cell line. Fluorescein (F)-labeled BSA co-localized with HGF on actin-rich cellular protrusions and with CM-DiI labeled pseudopodial plasma membranes. The F-BSA highlighted small, individual pseudopodial profiles more so than complex pseudopodial networks (reticulopodia) or unmigrated cells. Labeled human serum albumin also decorated pseudopodia preferentially. Albumin's association with pseudopodia may help to explain its selective accumulation in astrocytomas in vivo. The leaky BBB permits serum albumin to enter the microenvironment of astrocytomas thus allowing their invasive cells contact with serum albumin as a source of fatty acids that would be useful for remodeling cell membranes in pseudopodia. Thus, albumin potentially aids and marks invasion as it accumulates in these tumors.
Collapse
Affiliation(s)
- Marie E Beckner
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | | | | | |
Collapse
|
47
|
Abstract
Ovarian cancer is the sixth most common cancer and the fifth leading cause of cancer-related death among women in developed countries. Greater than 85% of human ovarian cancer arises within the ovarian surface epithelium (OSE), with the remainder derived from granulosa cells or, rarely, stroma or germ cells. The pathophysiology of ovarian cancer is the least understood among all major human malignancies because of a poor understanding of the aetiological factors and mechanisms of ovarian cancer progression. There is increasing evidence suggesting that several key reproductive hormones, such as GnRH, gonadotrophins and sex steroids, regulate the growth of normal OSE and ovarian cancer cells. The objective of this review was to highlight the effects of these endocrine factors on ovarian cancer cell growth and to summarize the signalling mechanisms involved in normal human OSE and its neoplastic counterparts.
Collapse
Affiliation(s)
- Peter C K Leung
- Department of Obstetrics and Gynecology, University of British Columbia, Child and Family Research Institute, Vancouver, British Columbia, Canada.
| | | |
Collapse
|
48
|
Simoncini T, Scorticati C, Mannella P, Fadiel A, Giretti MS, Fu XD, Baldacci C, Garibaldi S, Caruso A, Fornari L, Naftolin F, Genazzani AR. Estrogen receptor alpha interacts with Galpha13 to drive actin remodeling and endothelial cell migration via the RhoA/Rho kinase/moesin pathway. Mol Endocrinol 2006; 20:1756-71. [PMID: 16601072 DOI: 10.1210/me.2005-0259] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Sex steroids control cell movement and tissue organization; however, little is known of the involved mechanisms. This report describes the ongoing dynamic regulation by estrogen of the actin cytoskeleton and cell movement in human vascular endothelial cells that depends on rapid activation of the actin-regulatory protein moesin. Moesin activation is triggered by the interaction of the C-terminal portion of cell membrane estrogen receptor alpha with the G protein Galpha(13), leading to activation of the small GTPase RhoA and of the downstream effector Rho-associated kinase. The resulting phosphorylation of moesin on Thr(558) is the means of moesin's binding to actin and the remodeling of the actin cytoskeleton. This cascade of events ensues within minutes of estradiol administration and results in changes in cell morphology and to the development of specialized cell membrane structures such as ruffles and pseudopodia that are necessary for cell movement. These findings expand our knowledge of the basis of estrogen's effects on human cells, including the regulation of actin assembly, cell movement and migration. They highlight novel pathways of signal transduction of estrogen receptor alpha through nontranscriptional mechanisms. Furthermore, exposure of this estrogen receptor-dependent, nongenomic action of estrogen on human vascular endothelial cells is especially relevant to the present interest in the role of estrogen in cardiovascular protection.
Collapse
Affiliation(s)
- Tommaso Simoncini
- Molecular and Cellular Gynecological Endocrinology Laboratory, Department of Reproductive Medicine and Child Development, University of Pisa, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sarrió D, Rodríguez-Pinilla SM, Dotor A, Calero F, Hardisson D, Palacios J. Abnormal ezrin localization is associated with clinicopathological features in invasive breast carcinomas. Breast Cancer Res Treat 2006; 98:71-9. [PMID: 16538541 DOI: 10.1007/s10549-005-9133-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 12/04/2005] [Indexed: 01/26/2023]
Abstract
The membrane-cytoskeleton crosslinker ezrin is associated with malignant progression and metastasis in human neoplasias. To study the role of ezrin in breast cancer, we first assessed ezrin expression in a panel of breast cancer cell lines by western blot and confocal microscopy. Western blot revealed no differences in total ezrin levels among these breast cell lines. However, immunofluorescence staining revealed that Estrogen receptor (ER)-positive, noninvasive and nontumorigenic cell lines concentrated ezrin at the apical surface, whereas invasive cell lines localized ezrin in motile structures (membrane ruffles and filopodia) but also had more diffuse cytoplasmic staining. We next studied ezrin expression in 509 breast carcinomas using tissue microarrays. Immunohistochemical staining for ezrin, p53, Ki-67, phospho-Akt, HER2, and hormonal receptors was performed. Ezrin staining in normal breast epithelium localized at the apical, but not lateral, cell surface, whereas, in most breast tumor cases (331, 70.3%), it localized in the cytoplasm. Complete membranous staining occurred in 89 (18.9%) samples, and apical staining was seen in 51 (10.8%) cases. There were significant positive associations between cytoplasmic ezrin localization and adverse tumor characteristics such as high grade, high level of Ki-67 expression, hormonal-receptor negativity, and lymph-node metastases. Apical ezrin staining was associated with favorable clinicopathological features and node-negative tumors. Membranous ezrin staining was associated with high grade, strong HER2 and p-Akt expression. In conclusion, the switch of ezrin localization from the apical membrane to either the complete membrane or to the cytoplasm is correlated with dedifferentiation and adverse features in invasive breast tumors and cancer cell lines.
Collapse
Affiliation(s)
- David Sarrió
- Breast and Gynaecological Cancer Group, Molecular Pathology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | | | | | | | | | | |
Collapse
|
50
|
Syed V, Zhang X, Lau KM, Cheng R, Mukherjee K, Ho SM. Profiling estrogen-regulated gene expression changes in normal and malignant human ovarian surface epithelial cells. Oncogene 2005; 24:8128-43. [PMID: 16116479 DOI: 10.1038/sj.onc.1208959] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Estrogens regulate normal ovarian surface epithelium (OSE) cell functions but also affect epithelial ovarian cancer (OCa) development. Little is known about how estrogens play such opposing roles. Transcriptional profiling using a cDNA microarray containing 2400 named genes identified 155 genes whose expression was altered by estradiol-17beta (E2) in three immortalized normal human ovarian surface epithelial (HOSE) cell lines and 315 genes whose expression was affected by the hormone in three established OCa (OVCA) cell lines. All but 19 of the genes in these two sets were different. Among the 19 overlapping genes, five were found to show discordant responses between HOSE and OVCA cell lines. The five genes are those that encode clone 5.1 RNA-binding protein (RNPS1), erythrocyte adducin alpha subunit (ADD1), plexin A3 (PLXNA3 or the SEX gene), nuclear protein SkiP (SKIIP), and Rap-2 (rap-2). RNPS1, ADD1, rap-2, and SKIIP were upregulated by E2 in HOSE cells but downregulated by estrogen in OVCA cells, whereas PLXNA3 showed the reverse pattern of regulation. The estrogen effects was observed within 6-18 h of treatment. In silicon analyses revealed presence of estrogen response elements in the proximal promoters of all five genes. RNPS1, ADD1, and PLXNA3 were underexpressed in OVCA cell lines compared to HOSE cell lines, while the opposite was true for rap-2 and SKIIP. Functional studies showed that RNPS1 and ADD1 exerted multiple antitumor actions in OVCA cells, while PLXNA3 only inhibited cell invasiveness. In contrast, rap-2 was found to cause significant oncogenic effects in OVCA cells, while SKIIP promotes only anchorage-independent growth. In sum, gene profiling data reveal that (1) E2 exerts different actions on HOSE cells than on OVCA cells by affecting two distinct transcriptomes with few overlapping genes and (2) among the overlapping genes, a set of putative oncogenes/tumor suppressors have been identified due to their differential responses to E2 between the two cell types. These findings may explain the paradoxical roles of estrogens in regulating normal and malignant OSE cell functions.
Collapse
Affiliation(s)
- Viqar Syed
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | |
Collapse
|