51
|
Saddick SY. In vitro regulation of sheep ovarian surface epithelium (OSE) proliferation by local ovarian factors. Saudi J Biol Sci 2012; 19:285-90. [PMID: 23961188 DOI: 10.1016/j.sjbs.2012.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/21/2012] [Accepted: 03/22/2012] [Indexed: 11/26/2022] Open
Abstract
The ovarian surface epithelium (OSE) forms a lining around the entire ovary and actively participates in the ovulatory cycle. To investigate how specific growth factors and hormones affect OSE proliferation, the present study used sheep as a model to examine the effects of follicular and luteal products on the proliferation of sheep OSE cells in culture, and to analyse the influences of large antral follicles and corpora lutea (CL) on the expression of gonadotrophin receptors (FSHR and LHR) in the OSE. In the present study, follicular fluids from medium and large follicles, and extracts of corpora lutea stimulated the growth of OSE cells. The results of the present study showed that factors in the follicular fluid can induce OSE proliferative activity, and this stimulation effect could not be attributed to steroids in the follicular fluid since oestrogen and progesterone treatments failed to stimulate OSE cells. The expression of LH and FSH receptors over large follicles (5 mm or larger) was two and four times higher than those over stroma and CL, respectively. In conclusion, OSE proliferation in cycling sheep is associated with underlying developing follicles and CL, mediated by, at least in part, the up-regulation of gonadotrophin receptors, and facilitated by the action of mitogenic glycopeptides and growth factors, but not steroids.
Collapse
Affiliation(s)
- Salina Yahya Saddick
- Faculty of Science, Biology Department, King Abdulaziz University, Jeddah 21551, Saudi Arabia
| |
Collapse
|
52
|
Herr DR. Potential use of G protein-coupled receptor-blocking monoclonal antibodies as therapeutic agents for cancers. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 297:45-81. [PMID: 22608557 DOI: 10.1016/b978-0-12-394308-8.00002-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The therapeutic use of monoclonal antibodies (mAbs) is the fastest growing area of pharmaceutical development and has enjoyed significant clinical success since approval of the first mAb drug in1984. However, despite significant effort, there are still no approved therapeutic mAbs directed against the largest and most attractive family of drug targets: G protein-coupled receptors (GPCRs). GPCRs regulate essentially all cellular processes, including those that are fundamental to cancer pathology, such as proliferation, survival/drug resistance, migration, differentiation, tissue invasion, and angiogenesis. Many different GPCR isoforms are enhanced or dysregulated in multiple tumor types, and several GPCRs have known oncogenic activity. With approximately 350 distinct GPCRs in the genome, these receptors provide a rich landscape for the design of effective, targeted therapies for cancer, a uniquely heterogeneous disease family. While the generation of selective, efficacious mAbs has been problematic for these structurally complex integral membrane proteins, progress in the development of immunotherapeutics has been made by several independent groups. This chapter provides an overview of the roles of GPCRs in cancer and describes the current state of the art of GPCR-targeted mAb drugs.
Collapse
Affiliation(s)
- Deron R Herr
- Expression Drug Designs, LLC, San Marcos, California, USA
| |
Collapse
|
53
|
King SM, Burdette JE. Evaluating the progenitor cells of ovarian cancer: analysis of current animal models. BMB Rep 2011; 44:435-45. [PMID: 21777513 DOI: 10.5483/bmbrep.2011.44.7.435] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Serous ovarian cancer is one of the most lethal gynecological malignancies. Progress on effective diagnostics and therapeutics for this disease are hampered by ambiguity as to the cellular origins of this histotype of ovarian cancer, as well as limited suitable animal models to analyze early stages of disease. In this report, we will review current animal models with respect to the two proposed progenitor cells for serous ovarian cancer, the ovarian surface epithelium and the fallopian tube epithelium.
Collapse
Affiliation(s)
- Shelby M King
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 60607, USA
| | | |
Collapse
|
54
|
Cui J, Miner BM, Eldredge JB, Warrenfeltz SW, Dam P, Xu Y, Puett D. Regulation of gene expression in ovarian cancer cells by luteinizing hormone receptor expression and activation. BMC Cancer 2011; 11:280. [PMID: 21711548 PMCID: PMC3141782 DOI: 10.1186/1471-2407-11-280] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Accepted: 06/28/2011] [Indexed: 11/10/2022] Open
Abstract
Background Since a substantial percentage of ovarian cancers express gonadotropin receptors and are responsive to the relatively high concentrations of pituitary gonadotropins during the postmenopausal years, it has been suggested that receptor activation may contribute to the etiology and/or progression of the neoplasm. The goal of the present study was to develop a cell model to determine the impact of luteinizing hormone (LH) receptor (LHR) expression and LH-mediated LHR activation on gene expression and thus obtain insights into the mechanism of gonadotropin action on ovarian surface epithelial (OSE) carcinoma cells. Methods The human ovarian cancer cell line, SKOV-3, was stably transfected to express functional LHR and incubated with LH for various periods of time (0-20 hours). Transcriptomic profiling was performed on these cells to identify LHR expression/activation-dependent changes in gene expression levels and pathways by microarray and qRT-PCR analyses. Results Through comparative analysis on the LHR-transfected SKOV-3 cells exposed to LH, we observed the differential expression of 1,783 genes in response to LH treatment, among which five significant families were enriched, including those of growth factors, translation regulators, transporters, G-protein coupled receptors, and ligand-dependent nuclear receptors. The most highly induced early and intermediate responses were found to occupy a network impacting transcriptional regulation, cell growth, apoptosis, and multiple signaling transductions, giving indications of LH-induced apoptosis and cell growth inhibition through the significant changes in, for example, tumor necrosis factor, Jun and many others, supportive of the observed cell growth reduction in in vitro assays. However, other observations, e.g. the substantial up-regulation of the genes encoding the endothelin-1 subtype A receptor, stromal cell-derived factor 1, and insulin-like growth factor II, all of which are potential therapeutic targets, may reflect a positive mediation of ovarian cancer growth. Conclusion Overall, the present study elucidates the extensive transcriptomic changes of ovarian cancer cells in response to LH receptor activation, which provides a comprehensive and objective assessment for determining new cancer therapies and potential serum markers, of which over 100 are suggested.
Collapse
Affiliation(s)
- Juan Cui
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | | | | | | | | | | | | |
Collapse
|
55
|
Yoffou PH, Edjekouane L, Meunier L, Tremblay A, Provencher DM, Mes-Masson AM, Carmona E. Subtype specific elevated expression of hyaluronidase-1 (HYAL-1) in epithelial ovarian cancer. PLoS One 2011; 6:e20705. [PMID: 21695196 PMCID: PMC3112150 DOI: 10.1371/journal.pone.0020705] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 05/08/2011] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is morphologically heterogeneous being classified as serous, endometrioid, clear cell, or mucinous. Molecular genetic analysis has suggested a role for tumor suppressor genes located at chromosome 3p in serous EOC pathogenesis. Our objective was to evaluate the expression of HYAL1, located at chromosome 3p21.3, in these EOC subtypes, and to investigate its correlation with the expression of steroid hormone receptors. METHODOLOGY/PRINCIPAL FINDINGS We determined the mRNA expression of HYAL1, estrogen receptor (ER)-α, ERβ and progesterone receptor (PR) in EOC tumor samples and cell lines using quantitative RT-PCR. We also examined the expression of these genes in a publicly available microarray dataset. HYAL-1 enzyme activity was measured in EOC cell lines and in plasma samples from patients. We found that HYAL1 mRNA expression was elevated in clear cell and mucinous EOC tissue samples, but not in serous and endometrioid samples, normal ovaries or benign tumors. Similar results were obtained by two different techniques and with tissue sample cohorts from two independent institutions. Concordantly, HYAL1 mRNA levels and enzymatic activity were elevated only in EOC cell lines derived from clear cell and mucinous subtypes. We also showed that HYAL1 mRNA was inversely correlated to that of ERα specifically in clear cell and mucinous EOCs. Additionally, ectopic expression of ERα in a clear cell EOC cell line (ER- and PR-negative) induced 50% reduction of HYAL1 mRNA expression, supporting a role of ERα in HYAL1 gene regulation. Significantly, HYAL-1 activity was also high in the plasma of patients with these EOC subtypes. CONCLUSIONS/SIGNIFICANCE This is the first report showing high HYAL-1 levels in EOC and demonstrating HYAL1 gene repression by ERα. Our results identify Hyaluronidase-1 as a potential target/biomarker for clear cell and mucinous EOCs and especially in tumors with low ERα levels.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/blood
- Adenocarcinoma, Clear Cell/enzymology
- Adenocarcinoma, Clear Cell/pathology
- Adenocarcinoma, Mucinous/blood
- Adenocarcinoma, Mucinous/enzymology
- Adenocarcinoma, Mucinous/pathology
- Biomarkers, Tumor/blood
- Carcinoma, Ovarian Epithelial
- Cell Line, Tumor
- Culture Media, Conditioned
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hyaluronoglucosaminidase/genetics
- Hyaluronoglucosaminidase/metabolism
- Neoplasms, Glandular and Epithelial/blood
- Neoplasms, Glandular and Epithelial/classification
- Neoplasms, Glandular and Epithelial/enzymology
- Neoplasms, Glandular and Epithelial/genetics
- Ovarian Neoplasms/blood
- Ovarian Neoplasms/classification
- Ovarian Neoplasms/enzymology
- Ovarian Neoplasms/genetics
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
Collapse
Affiliation(s)
| | - Lydia Edjekouane
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada
| | - Liliane Meunier
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - André Tremblay
- Department of Obstetrics and Gynecology, University of Montreal, Montreal, Quebec, Canada
- Department of Biochemistry, University of Montreal, Montreal, Quebec, Canada
- Research Center, Centre hospitalier universitaire Ste-Justine, Montreal, Quebec, Canada
| | - Diane Michèle Provencher
- Department of Obstetrics and Gynecology, University of Montreal, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Anne-Marie Mes-Masson
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Department of Obstetrics and Gynecology, University of Montreal, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Euridice Carmona
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
56
|
Wright JW, Jurevic L, Stouffer RL. Dynamics of the primate ovarian surface epithelium during the ovulatory menstrual cycle. Hum Reprod 2011; 26:1408-21. [PMID: 21421660 DOI: 10.1093/humrep/der057] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) risk correlates strongly with the number of ovulations that a woman experiences. The primary source of EOC in women is the ovarian surface epithelium (OSE). Mechanistic studies on the etiology of OSE transformation to EOC cannot be realistically performed in women. Selecting a suitable animal model to investigate the normal OSE in the context of ovulation should be guided by the model's reproductive similarities to women in natural features that are thought to contribute to EOC risk. METHODS We selected the non-human primate, rhesus macaque, as a surrogate to study the normal OSE during the natural menstrual cycle. We investigated OSE morphology and marker expression, plus cell proliferation and death in relation to menstrual cycle stage and ovulation. RESULTS OSE cells displayed a morphological range from squamous to columnar. Cycle-independent parameters and cycle-dependent changes were observed for OSE histology, steroid receptor expression, cell death, DNA repair and cell adhesion. Contrary to findings in non-primates, primate OSE cells were not manifestly cleared from the site of ovulation, nor were proliferation rates affected by ovulation or stage of the menstrual cycle. DNA repair proteins were more highly expressed in OSE than in other ovarian cells. CONCLUSIONS This study identifies significant differences between primate and non-primate OSE. In contrast to established views, ovulation-induced death and proliferation are not indicated as prominent contributors to EOC risk, but disruption of OSE cadherin-mediated adhesion may be, as could the loss of ovary-mediated chronic suppression of proliferation and elevation of DNA repair potential.
Collapse
Affiliation(s)
- Jay W Wright
- Division of Reproductive Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA.
| | | | | |
Collapse
|
57
|
Puett D, Angelova K, da Costa MR, Warrenfeltz SW, Fanelli F. The luteinizing hormone receptor: insights into structure-function relationships and hormone-receptor-mediated changes in gene expression in ovarian cancer cells. Mol Cell Endocrinol 2010; 329:47-55. [PMID: 20444430 PMCID: PMC2946427 DOI: 10.1016/j.mce.2010.04.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 04/09/2010] [Accepted: 04/26/2010] [Indexed: 10/19/2022]
Abstract
The luteinizing hormone receptor (LHR), one of the three glycoprotein hormone receptors, is necessary for critical reproductive processes, including gonadal steroidogenesis, oocyte maturation and ovulation, and male sex differentiation. Moreover, it has been postulated to contribute to certain neoplasms, particularly ovarian cancer. A member of the G protein-coupled receptor family, LHR contains a relatively large extracellular domain responsible for high affinity hormone binding; transmembrane activation then leads to G protein coupling and subsequent second messenger production. This review deals with recent advances in our understanding of LHR structure and structure-function relationships, as well as hormone-mediated changes in gene expression in ovarian cancer cells expressing LHR. Suggestions are also made for critical gaps that need to be filled as the field advances, including determination of the three-dimensional structure of inactive and active receptor, elucidation of the mechanism by which hormone binding to the extracellular domain triggers the activation of Gs, clarification of the putative roles of LHR in non-gonadal tissues, and the role, if any, of activated receptor in the development or progression of ovarian cancer.
Collapse
Affiliation(s)
- David Puett
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA.
| | | | | | | | | |
Collapse
|
58
|
Huhtaniemi I. Are gonadotrophins tumorigenic--a critical review of clinical and experimental data. Mol Cell Endocrinol 2010; 329:56-61. [PMID: 20471448 DOI: 10.1016/j.mce.2010.04.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 04/12/2010] [Accepted: 04/26/2010] [Indexed: 01/17/2023]
Abstract
The growth of many gonadal and extragonadal tumors is stimulated by gonadal sex hormones. Because gonadal hormone production is regulated by pituitary gonadotrophins, the latter hormones can be considered as indirect tumor promoters. In addition, there is a growing body of evidence that both gonadal (e.g. ovarian cancer) and extragonadal (e.g. breast, uterus, prostate and adrenal) tumors express gonadotrophin receptors, indicating the possibility of a direct tumorigenic role for FSH and LH. The purpose of this brief review is to present a critical evaluation of the current information, both clinical and experimental, about the direct involvement of gonadotrophins in the induction and growth of gonadal and extragonadal tumors.
Collapse
Affiliation(s)
- Ilpo Huhtaniemi
- Department of Surgery and Cancer, IRDB Building, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
59
|
Pérez-López FR, Chedraui P, Troyano-Luque JM. Peri- and post-menopausal incidental adnexal masses and the risk of sporadic ovarian malignancy: new insights and clinical management. Gynecol Endocrinol 2010; 26:631-43. [PMID: 20500106 DOI: 10.3109/09513590.2010.487611] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Adnexal masses are common among peri- and post-menopausal women. Although ovarian cancer is a significant cause of mortality in menopausal women, large population-based studies demonstrate that the majority of adnexal masses are benign. Despite this, the appearance of an adnexal mass is a concern for the patient and an insight exercise for physicians. In most cases, an adnexal enlargement is an incidental finding, generally corresponding to a benign cyst and easily diagnosed by conventional ultrasound. Exceptionally an ovarian tumour may be malignant and should be treated as early as possible. When conventional ultrasound renders complex morphology other diagnostic tools must be used such as: colour Doppler and functional tumour vessel properties, serum CA 125 levels, nuclear magnetic resonance imaging and in some cases laparoscopy. Several new tumour markers are being studied for clinical application, although there are presently no clear recommendations. Adnexal masses with benign morphological and functional properties must be periodically monitored as an alternative to surgery since malignant transformation is exceptional.
Collapse
Affiliation(s)
- Faustino R Pérez-López
- Department of Obstetrics and Gynaecology, Facultad de Medicina, Universidad de Zaragoza, Hospital Clínico de Zaragoza, Zaragoza 50009, Spain.
| | | | | |
Collapse
|
60
|
Mancinelli R, Onori P, DeMorrow S, Francis H, Glaser S, Franchitto A, Carpino G, Alpini G, Gaudio E. Role of sex hormones in the modulation of cholangiocyte function. World J Gastrointest Pathophysiol 2010; 1:50-62. [PMID: 21607142 PMCID: PMC3097944 DOI: 10.4291/wjgp.v1.i2.50] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 05/28/2010] [Accepted: 06/04/2010] [Indexed: 02/06/2023] Open
Abstract
Over the last years, cholangiocytes, the cells that line the biliary tree, have been considered an important object of study for their biological properties which involves bile formation, proliferation, injury repair, fibrosis and angiogenesis. Cholangiocyte proliferation occurs in all pathologic conditions of liver injury where it is associated with inflammation and regeneration. During these processes, biliary cells start to secrete different cytokines, growth factors, neuropeptides and hormones which represent potential mechanisms for cross talk with other liver cells. Several studies suggest that hormones, and in particular, sex hormones, play a fundamental role in the modulation of the growth of this compartment in the injured liver which functionally conditions the progression of liver disease. Understanding the mechanisms of action and the intracellular pathways of these compounds on cholangiocyte pathophysiology will provide new potential strategies for the management of chronic liver diseases. The purpose of this review is to summarize the recent findings on the role of sex hormones in cholangiocyte proliferation and biology.
Collapse
|
61
|
Peritoneal adhesion and angiogenesis in ovarian carcinoma are inversely regulated by hyaluronan: the role of gonadotropins. Neoplasia 2010; 12:51-60. [PMID: 20072653 DOI: 10.1593/neo.91272] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2009] [Revised: 10/30/2009] [Accepted: 10/30/2009] [Indexed: 11/18/2022]
Abstract
Ovarian carcinoma is the leading cause of death among gynecologic cancers. Although transformation of the outer ovarian epithelium was linked with ovulation, the disease is significantly more prevalent and severe in postmenopausal women. We postulated that menopause could augment ovarian cancer progression through the effects of gonadotropins on multifocal seeding to the mesothelial layer lining the peritoneum. This seeding is mediated by integrins as well as by CD44 interaction with hyaluronan (HA). Here, we report the effect of gonadotropins on HA synthesis and degradation and on peritoneal adhesion. A significant concentration- and time-dependent induction in expression levels of HA synthases (HASs) and hyaluronidases (Hyals) was observed in vitro on stimulation of human epithelial ovarian carcinoma cells by gonadotropins. Hormonal regulation of HA-mediated adhesion was manifested in vivo as well, by fluorescence microscopy of stained MLS multicellular tumor spheroids. The number of spheroids adhered to the mesothelium of ovariectomized CD-1 nude mice 9.5 hours after intraperitoneal insertion was significantly higher than in nonovariectomized mice. Inhibition of HA synthesis by 6-diazo-5-oxo-1-norleucine (DON) both in spheroids and ovariectomized mice significantly reduced the number of adhered spheroids. Thus, the change in the hormonal environment during menopause assists in HA-dependent adherence of ovarian cancer spheroids onto the peritoneum. However, HA is antiangiogenic and it can significantly suppress tumor progression. Accordingly, angiogenesis of the adhered spheroids was significantly elevated in DON-treated tumors. These results can explain the selective pressure that can lead to simultaneously increased tumor expression of both HASs and Hyals.
Collapse
|
62
|
Sapoznik S, Cohen B, Tzuman Y, Meir G, Ben-Dor S, Harmelin A, Neeman M. Gonadotropin-regulated lymphangiogenesis in ovarian cancer is mediated by LEDGF-induced expression of VEGF-C. Cancer Res 2010; 69:9306-14. [PMID: 19934313 DOI: 10.1158/0008-5472.can-09-1213] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The risk and severity of ovarian carcinoma, the leading cause of gynecologic malignancy death, are significantly elevated in postmenopausal women. Ovarian failure at menopause, associated with a reduction in estrogen secretion, results in an increase of the gonadotropic luteinizing hormone (LH) and follicle-stimulating hormone (FSH), suggesting a role for these hormones in facilitating the progression of ovarian carcinoma. The current study examined the influence of hormonal stimulation on lymphangiogenesis in ovarian cancer cells. In vitro stimulation of ES2 ovarian carcinoma cells with LH and FSH induced expression of vascular endothelial growth factor (VEGF)-C. In vivo, ovariectomy of mice resulted in activation of the VEGF-C promoter in ovarian carcinoma xenografts, increased VEGF-C mRNA level, and enhanced tumor lymphangiogenesis and angiogenesis. Seeking the molecular mechanism, we examined the role of lens epithelium-derived growth factor (LEDGF/p75) and the possible contribution of its putative target, a conserved stress-response element identified in silico in the VEGF-C promoter. Using chromatin immunoprecipitation, we showed that LEDGF/p75 indeed binds the VEGF-C promoter, and binding is augmented by FSH. A corresponding hormonally regulated increase in the LEDGF/p75 mRNA and protein levels was observed. Suppression of LEDGF/p75 expression using small interfering RNA, suppression of LH and FSH production using the gonadotropin-releasing hormone antagonist cetrorelix, or mutation of the conserved stress-response element suppressed the hormonally induced expression of VEGF-C. Overall, our data suggest a possible role for elevated gonadotropins in augmenting ovarian tumor lymphangiogenesis in postmenopausal women.
Collapse
Affiliation(s)
- Stav Sapoznik
- Department of Biological Regulation, Weizmann Institute, Rehovot, Israel
| | | | | | | | | | | | | |
Collapse
|
63
|
Serin IS, Tanriverdi F, Ata CD, Akalin H, Ozcelik B, Ozkul Y, Kelestimur F. GnRH-II mRNA expression in tumor tissue and peripheral blood mononuclear cells (PBMCs) in patients with malignant and benign ovarian tumors. Eur J Obstet Gynecol Reprod Biol 2009; 149:92-6. [PMID: 20018426 DOI: 10.1016/j.ejogrb.2009.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 09/15/2009] [Accepted: 11/16/2009] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate the expression of the second form of GnRH (GnRH-II) in tumor tissue and peripheral blood mononuclear cells (PBMCs) in malignant and benign ovarian tumors in humans. STUDY DESIGN Sixty-six women were studied: 24 with epithelial ovarian carcinomas, 22 with benign ovarian tumors and 20 in the control group undergoing surgery. Malignant, benign and normal ovarian tissue and PBMCs were obtained for measurement of GnRH-II mRNA levels using quantitative real-time RT-PCR. RESULT(S) The expression of GnRH-II was found to be 1.5 times higher in malignant ovarian tumors compared with benign ovarian tumors and the control group in post-menopausal patients (P<0.01). In the post-menopausal patient group with malignant ovarian tumors, there were significant positive correlations between serum FSH level and ovarian tissue GnRH-II mRNA expression (r=0.68; P=0.03), and serum LH level and ovarian tissue GnRH-II mRNA expression (r=0.71; P=0.02). Controls, benign and malignant groups were similar in terms of GnRH-II expression in PBMCs in the pre- and post-menopausal periods. There was no significant correlation between ovarian tissue GnRH-II mRNA expression vs. PBMC GnRH-II mRNA expression in patient and control groups. CONCLUSION(S) We have shown increased GnRH-II expression in human ovarian cancer tissue in post-menopausal women in vivo. Expression of GnRH-II in PBMCs did not reflect the local GnRH-II expression levels in ovarian tissue. These preliminary data suggest that local GnRH-II may participate in the regulation of ovarian tumor growth in post-menopausal women.
Collapse
Affiliation(s)
- Ibrahim Serdar Serin
- Department of Obstetrics and Gynecology, Erciyes University, 38039 Kayseri, Turkey.
| | | | | | | | | | | | | |
Collapse
|
64
|
Zhang XY, Chen J, Zheng YF, Gao XL, Kang Y, Liu JC, Cheng MJ, Sun H, Xu CJ. Follicle-stimulating hormone peptide can facilitate paclitaxel nanoparticles to target ovarian carcinoma in vivo. Cancer Res 2009; 69:6506-14. [PMID: 19638590 DOI: 10.1158/0008-5472.can-08-4721] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chemotherapy is an important treatment for ovarian cancer. However, conventional chemotherapy has inevitable drawbacks due to side effects from nonspecific biodistribution of the chemotherapeutic drugs. To solve such problem, targeted delivery approaches were developed. The targeted delivery approaches combine drug carriers with the targeting system and can preferentially bring drugs to the targeted sites. Follicle-stimulating hormone receptor (FSHR) is an ovarian cancer-specific receptor. By using a peptide derived from FSH (amino acids 33-53 of the FSH beta chain, named as FSH33), we developed a conjugated nanoparticle, FSH33-NP, to target FSHR in ovarian cancer. FSH33-NP was tested for recognition specificity and uptake efficiency on FSHR-expressing cells. Then, the antitumor efficiency of paclitaxel (PTX)-loaded FSH33-NP (FSH33-NP-PTX) was determined. FSH33-NP-PTX displayed stronger antiproliferation and antitumor effects compared with free PTX or naked PTX-loaded nanoparticles (NP-PTX) both in vitro and in vivo. In summary, this novel FSH33-NP delivery system showed very high selectivity and efficacy for FSHR-expressing tumor tissues. Therefore, it has good potential to become a new therapeutic approach for patients with ovarian cancer.
Collapse
Affiliation(s)
- Xiao-yan Zhang
- Obstetrics and Gynecology Hospital, Fudan University, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Warrenfeltz SW, Lott SA, Palmer TM, Gray JC, Puett D. Luteinizing hormone-induced up-regulation of ErbB-2 is insufficient stimulant of growth and invasion in ovarian cancer cells. Mol Cancer Res 2009; 6:1775-85. [PMID: 19010824 DOI: 10.1158/1541-7786.mcr-08-0214] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The effects of luteinizing hormone (LH), a gonadotropic hormone implicated in the development of ovarian cancer, are mediated by specific binding to its G protein-coupled receptor, the LH receptor (LHR). Activated LHR initiates second messenger responses, including cyclic AMP (cAMP) and inositol phosphate. Because cAMP increases expression of ErbB-2, a receptor tyrosine kinase whose overexpression in cancers correlates with poor survival, we hypothesized that LH may regulate ErbB-2 expression. Cell surface LHR expression in stable transformants of the ErbB-2-overexpressing ovarian cancer cell line SKOV3 was confirmed by PCR and whole-cell ligand binding studies. Second messenger accumulation in the LHR-expressing cells confirmed signaling through Gs and Gq. Western blots of total protein revealed that LHR introduction up-regulated ErbB-2 protein expression 2-fold and this was further up-regulated in a time- and dose-dependent manner in response to LH. Forskolin and 8Br-cAMP also up-regulated ErbB-2 in both LHR-expressing and mock-transfected cells, indicating that regulation of ErbB-2 is a cAMP-mediated event. Kinase inhibitor studies indicated the involvement of protein kinase A-mediated, protein kinase C-mediated, epidermal growth factor receptor-mediated, and ErbB-2-mediated mechanisms. The LH-induced up-regulation of ErbB-2 was insufficient to overcome the negative effects of LH on proliferation, invasion, and migration. A molecular signature for this nonaggressive phenotype was determined by Taqman array to include increased and decreased expression of genes encoding adhesion proteins and metalloproteinases, respectively. These data establish a role for LH and LHR in the regulation of ErbB-2 expression and suggest that, in some systems, ErbB-2 up-regulation alone is insufficient in producing a more aggressive phenotype.
Collapse
Affiliation(s)
- Susanne W Warrenfeltz
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | | | | | | | | |
Collapse
|
66
|
Zhang Z, Jia L, Feng Y, Zheng W. Overexpression of follicle-stimulating hormone receptor facilitates the development of ovarian epithelial cancer. Cancer Lett 2009; 278:56-64. [PMID: 19181441 DOI: 10.1016/j.canlet.2008.12.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2008] [Revised: 11/24/2008] [Accepted: 12/17/2008] [Indexed: 11/25/2022]
Abstract
We previously showed that the expressing level of FSH receptor (FSHR) increased from ovarian epithelial inclusions (OEIs) to benign ovarian epithelial tumors (OETs) and to borderline OETs, whereas FSHR levels decreased with an increase in carcinoma grade. The aim of this study was to investigate the role of FSHR in OET development. MCV152 cells with FSHR overexpression showed an increased cellular proliferation and invasive capacity, which was associated with reduced levels of prohibitin and RII-beta expression and increased levels of HER-2/neu, c-Myc, and EGFR expression. Overexpression of FSHR may be associated with an elevated level of OET cell proliferation via an enhanced activity of potential oncogenic pathways. Therefore, the findings in this study suggest that overexpression of FSHR may play a role in OET development.
Collapse
Affiliation(s)
- Zhenbo Zhang
- Hospital of Obstetrics and Gynecology, Shanghai Medical College Fudan University, 379 Fang Xie Road, Shanghai 200011, China
| | | | | | | |
Collapse
|
67
|
The haplotype of two FSHR polymorphisms in ovarian cancer--a potential role of ethnology in risk modification. Gynecol Oncol 2009; 112:486-9. [PMID: 19147210 DOI: 10.1016/j.ygyno.2008.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 11/25/2008] [Accepted: 12/09/2008] [Indexed: 11/20/2022]
Abstract
OBJECTIVES The precise role of gonadotropins in the carcinogenesis of epithelial ovarian cancer remains uncertain. Recently, the haplotype of two single nucleotide polymorphisms, Thr307Ala (rs6165) and Asn680Ser (rs6166), has been described as a risk factor for ovarian cancer in Chinese women. In this study we investigated the impact of this haplotype regarding the risk to develop ovarian cancer as well as possible effects upon the clinical course in a Caucasian patient sample. SUBJECTS AND METHODS Determination of genotypes in 115 patients with primary epithelial ovarian cancer and 115 age-matched controls was performed by Pyrosequencing for Thr307Ala and by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) technique for Asn680Ser. RESULTS Analysis of the genotypes revealed almost complete linkage disequilibrium of both SNPs. The distribution of genotypes was not statistically significant different between ovarian cancer patients and age-matched controls. Clinical parameters such as overall survival, CA12-5 elevation at primary diagnosis, age at diagnosis, FIGO stage, grading, and platinum resistance were not statistically significantly different regarding genotypes. CONCLUSIONS We could not confirm the FSHR Ala307-Ser680 haplotype as a risk factor for epithelial ovarian cancer in Caucasian women. Hence, the modification of tumor risk may be affected by the ethnology of the patient collective. We could not find any associations of clinical parameters or course of the disease with the different genotypes.
Collapse
|
68
|
Doyle LK, Donadeu FX. Regulation of the proliferative activity of ovarian surface epithelial cells by follicular fluid. Anim Reprod Sci 2008; 114:443-8. [PMID: 19042098 DOI: 10.1016/j.anireprosci.2008.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 10/13/2008] [Accepted: 10/17/2008] [Indexed: 11/29/2022]
Abstract
Despite critical roles of the ovarian surface epithelium (OSE) in ovulation and post-ovulatory wound repair, little is known about the physiological mechanism regulating OSE proliferation. A role of follicles and corpora lutea in locally regulating the proliferative activity of OSE has been suggested. In this study, the effects of follicular and luteal products on proliferation of cultured OSE cells were tested using cells obtained from seasonally anoestrous ewes. Follicular fluid but not luteal extracts induced OSE cell proliferation (2.5-fold relative to untreated controls; P<0.0001). The response of OSE cells was not affected by follicle size or previous charcoal-extraction of follicular fluid (P>0.1). Treatment with IGF-1 (2.2-fold; P<0.01), EGF (1.9-fold; P<0.01) and, to a lesser extent, FSH (P<0.05) also induced OSE cell proliferation. In contrast, oestradiol or progesterone did not induce cell proliferation or enhance the effects of FSH on proliferation (P>0.1). It was concluded that follicular fluid can directly stimulate ovine OSE cell proliferation and that this effect is attributable to non-steroidal mitogens.
Collapse
Affiliation(s)
- L K Doyle
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Roslin BioCentre, Midlothian EH25 9PS, UK
| | | |
Collapse
|
69
|
So WK, Cheng JC, Poon SL, Leung PCK. Gonadotropin-releasing hormone and ovarian cancer: a functional and mechanistic overview. FEBS J 2008; 275:5496-511. [DOI: 10.1111/j.1742-4658.2008.06679.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
70
|
Huang Y, Hua K, Zhou X, Jin H, Chen X, Lu X, Yu Y, Zha X, Feng Y. Activation of the PI3K/AKT pathway mediates FSH-stimulated VEGF expression in ovarian serous cystadenocarcinoma. Cell Res 2008; 18:780-91. [DOI: 10.1038/cr.2008.70] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
71
|
Chen X, Aravindakshan J, Yang Y, Sairam MR. Early alterations in ovarian surface epithelial cells and induction of ovarian epithelial tumors triggered by loss of FSH receptor. Neoplasia 2007; 9:521-31. [PMID: 17603635 PMCID: PMC1899255 DOI: 10.1593/neo.07238] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 05/01/2007] [Accepted: 05/02/2007] [Indexed: 11/18/2022]
Abstract
Little is known about the behavior of the ovarian surface epithelium (OSE), which plays a central role in ovarian cancer etiology. It has been suggested that incessant ovulation causes OSE changes leading to transformation and that high gonadotropin levels during postmenopause activate OSE receptors, inducing proliferation. We examined the chronology of OSE changes, including tumor appearance, in a mouse model where ovulation never occurs due to deletion of follitropin receptor. Changes in epithelial cells were marked by pan-cytokeratin (CK) staining. Histologic changes and CK staining in the OSE increased from postnatal day 2. CK staining was observed inside the ovary by 24 days and increased thereafter in tumor-bearing animals. Ovaries from a third of aged (1 year) mutant mice showed CK deep inside, indicating cell migration. These tumors resembled serous papillary adenoma of human ovaries. Weak expression of GATA-4 and elevation of PCNA, cyclooxygenase-1, cyclooxygenase-2, and platelet-derived growth factor receptors alpha and beta in mutants indicated differences in cell proliferation, differentiation, and inflammation. Thus, we report that OSE changes occur long before epithelial tumors appear in FORKO mice. Our results suggest that neither incessant ovulation nor follicle-stimulating hormone receptor presence in the OSE is required for inducing ovarian tumors; thus, other mechanisms must contribute to ovarian tumorigenesis.
Collapse
Affiliation(s)
- Xinlei Chen
- Molecular Reproduction Research Laboratory, Clinical Research Institute of Montreal, Univerisité de Montréal, Montreal, Quebec H2W 1R7, Canada
| | | | | | | |
Collapse
|
72
|
Abstract
Ovarian epithelial cancer (OEC) accounts for 90% of all ovarian cancers and is the leading cause of death from gynecological cancers in North America and Europe. Despite its clinical significance, the factors that regulate the development and progression of ovarian cancer are among the least understood of all major human malignancies. The two gonadotropins, FSH and LH, are key regulators of ovarian cell functions, and the potential role of gonadotropins in the pathogenesis of ovarian cancer is suggested. Ovarian carcinomas have been found to express specific receptors for gonadotropins. The presence of gonadotropins in ovarian tumor fluid suggests the importance of these factors in the transformation and progression of ovarian cancers as well as being prognostic indicators. Functionally, there is evidence showing a direct action of gonadotropins on ovarian tumor cell growth. This review summarizes the key findings and recent advances in our understanding of these peptide hormones in ovarian cancer development and progression and their role in potential future cancer therapy. We will first discuss the supporting evidence and controversies in the "gonadotropin theory" and the use of animal models for exploring the involvement of gonadotropins in the etiology of ovarian cancer. The role of gonadotropins in regulating the proliferation, survival, and metastasis of OEC is next summarized. Relevant data from ovarian surface epithelium, which is widely believed to be the precursor of OEC, are also described. Finally, we will discuss the clinical applications of gonadotropins in ovarian cancer and the recent progress in drug development.
Collapse
Affiliation(s)
- Jung-Hye Choi
- Department of Obstetrics and Gynecology, Zhejiang University School of Medicine, China
| | | | | | | |
Collapse
|
73
|
Schumacher JJ, Dings RPM, Cosin J, Subramanian IV, Auersperg N, Ramakrishnan S. Modulation of angiogenic phenotype alters tumorigenicity in rat ovarian epithelial cells. Cancer Res 2007; 67:3683-90. [PMID: 17440080 DOI: 10.1158/0008-5472.can-06-3608] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascular endothelial growth factor (VEGF) expression correlates with microvessel density, stage, malignant ascites, metastasis, and survival in ovarian cancer. By transducing VEGF165 into a nontumorigenic rat ovarian surface epithelial cell line (ROSE199), we investigated the direct effect of an angiogenic phenotype on tumor development. The neu oncogene, which is overexpressed in >30% of ovarian cancers, was used in comparison. Neu-transfected ROSE199 cells showed phenotypic characteristics of transformation in vitro with an abundance of focus-forming units in monolayer cultures and anchorage-independent growth in soft agar. In contrast, VEGF-secreting ROSE199 cells (VR) retained normal morphology and in vitro growth characteristics (e.g., proliferation rate) compared with parental ROSE199 cells. Interestingly, injection of VR cells into athymic mice formed malignant ascites in 100% of the animals when injected into the peritoneum and developed vascularized tumors in 85% of the mice when injected s.c. Furthermore, blocking VEGF-mediated signaling by the Flk-1/KDR receptor kinase inhibitor SU5416 significantly inhibited the growth of VR tumors. To validate that the proangiogenic switch is responsible for tumor development, the angiogenic phenotype was balanced by the inducible coexpression of endostatin under the control of Tet-activated promoter. Coexpression of endostatin along with VEGF reversed the tumorigenic phenotype of VR cells. These studies show that alterations in the angiogenic characteristics of ovarian surface epithelium may play an important role in the etiology of ovarian cancer, and that inhibition of angiogenesis can be effective in the treatment of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Jennifer J Schumacher
- Department of Pharmacology, University of Minnesota, 321 Church Street SE, Minneapolis, MN 55445, USA
| | | | | | | | | | | |
Collapse
|
74
|
Abstract
Gonadotropins, follicle-stimulating hormone and luteinizing hormone are key regulators in ovarian function, acting in an endocrine manner to regulate gametogenesis and steroidogenesis. In addition to normal tissue, gonadotropin receptors have also been demonstrated in ovarian carcinoma cell lines and primary tumors, suggesting that the gonadotropins may play a role in the pathophysiology of ovarian cancer. Thus, understanding mechanisms involved in signaling transduction by the gonadotropin receptors are of considerable interest and potential significance. In the ovary, gonadotropins initiate their cellular responses by binding to their G-protein-coupled receptors and activation of specific downstream intracellular effectors and signal pathways, including those of protein kinases A and C and mitogen-activated protein kinase. Recently, gonadotropins were shown to stimulate nuclear accumulation of β-catenin, which controls lymphoid-enhancing factor/T-cell factor family-sensitive gene expression. β-catenin has a pivotal function in the control of cell fate. The ability of gonadotropins to regulate β-catenin provides a new dimension of knowledge linking pituitary hormones to the β-catenin signaling in normal ovarian physiology and demonstrating how its dysregulation can contribute to the development of ovarian cancer.
Collapse
Affiliation(s)
- Yuen-Lam Pon
- a University of Hong Kong, Department of Zoology, Pokfulam Road, Hong Kong.
| | - Alice St Wong
- b Assistant Professor, University of Hong Kong, Department of Zoology, 4S-14 Kadoorie Biological Sciences Building, Pokfulam Road, Hong Kong.
| |
Collapse
|
75
|
Fleming JS, McQuillan HJ, Millier MJ, Beaugié CR, Livingstone V. E-cadherin expression and bromodeoxyuridine incorporation during development of ovarian inclusion cysts in age-matched breeder and incessantly ovulated CD-1 mice. Reprod Biol Endocrinol 2007; 5:14. [PMID: 17425809 PMCID: PMC1855058 DOI: 10.1186/1477-7827-5-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Accepted: 04/11/2007] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Female CD-1/Swiss Webster mice subjected to incessant ovulation for 8 months and 12-month breeder mice both developed ovarian inclusion cysts similar to serous cystadenomas. The majority of cysts appeared to be dilated rete ovarii tubules, but high ovulation number resulted in more cortical inclusion cysts. We hypothesized that comparison of inclusion cyst pathology in animals of the same age, but with differences in total lifetime ovulation number, might allow us to determine distinguishing characteristics of the two types of cyst. METHODS Ovaries from breeder mice (BR) or females subjected to incessant ovulation (IO) were compared at 6-, 9- and 12-months of age. Ovaries were serially sectioned and cysts characterized with regard to location and histology, E-cadherin immunoreactivity and rates of BrdU incorporation. RESULTS Inclusion cysts developed with age in BR and IO ovaries. The majority of cysts were connected to the ovarian hilus. Two cortical inclusion cysts were observed in ten IO ovaries and one in ten BR ovaries. Low or no E-cadherin immuno-staining was seen in the OSE of all mice studied. Conversely, strong membrane immuno-staining was observed in rete ovarii epithelial cells. Variable E-cadherin immunoreactivity was seen in cells of hilar inclusion cysts, with strong staining observed in cuboidal ciliated cells and little or no staining in flat epithelial cells. Two of the three cortical cysts contained papillae, which showed E-cadherin immuno-staining at the edge of cells. However hilar and cortical cysts were not distinguishable by morphology, cell type or E-cadherin immunoreactivity. BrdU incorporation in cyst cells (1.4% [95% CI: 1.0 to 2.1]) was greater than in OSE (0.7% [95% CI: 0.4 to 1.2]) and very few BrdU-labeled cells were observed in rete ovarii at any age. Incessant ovulation significantly increased BrdU incorporation in OSE of older animals. CONCLUSION These experiments confirm ovarian inclusion cysts develop with age in the CD-1 mouse strain, irrespective of total ovulation burden. We conclude longer periods of incessant ovulation do not lead to significant changes in inclusion cyst formation or steroidogenesis in CD-1 mice and inclusion cyst type can not be distinguished by morphology, cell proliferation rate or E-cadherin immunoreactivity.
Collapse
Affiliation(s)
- Jean S Fleming
- Eskitis Institute of Cell & Molecular Therapies, Griffith University Nathan campus, Nathan, QLD 4111, Australia
| | - H James McQuillan
- Department of Anatomy and Structural Biology, University of Otago School of Medical Sciences, PO Box 913, Dunedin, New Zealand
| | - Melanie J Millier
- Department of Anatomy and Structural Biology, University of Otago School of Medical Sciences, PO Box 913, Dunedin, New Zealand
| | - Clare R Beaugié
- Department of Anatomy and Structural Biology, University of Otago School of Medical Sciences, PO Box 913, Dunedin, New Zealand
| | - Vicki Livingstone
- Department of Preventive & Social Medicine, University of Otago Health Sciences, PO Box 913, Dunedin, New Zealand
| |
Collapse
|
76
|
Li Y, Ganta S, Cheng C, Craig R, Ganta RR, Freeman LC. FSH stimulates ovarian cancer cell growth by action on growth factor variant receptor. Mol Cell Endocrinol 2007; 267:26-37. [PMID: 17234334 PMCID: PMC1880879 DOI: 10.1016/j.mce.2006.11.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 11/28/2006] [Indexed: 10/23/2022]
Abstract
A number of FSH receptor (FSH-R) isoforms with distinct structural motifs and signaling paradigms have been described, including a single transmembrane domain variant that functions as a growth factor type receptor (FSH-R3). This study tested the hypothesis that FSH can stimulate ovarian cancer cell proliferation by acting on FSH-R3, using the tumorigenic mouse ovarian surface epithelial cell (MOSEC) line ID8. FSH enhanced ID8 proliferation in a concentration-dependent fashion. Moreover, FSH-treatment of ID8 elicited intracellular events consistent with activation of FSH-R3 and distinct from those associated with activation of the canonical G-protein coupled FSH-R isoform (FSH-R1). Specifically, the FSH-R3 signaling pathway included cAMP-independent activation of ERK downstream of an SNX-482 sensitive component likely to be the Cav2.3 calcium channel. Northern analysis using probes specific for exons 7 and 11 of FSH-R identified consistently only one 1.9kb transcript. Immunoblot analysis confirmed expression of FSH-R3 but not FSHR-1 in ID8. Together, these data suggest that FSH-R3 signaling promotes proliferation of ovarian cancer cells.
Collapse
Affiliation(s)
- Y Li
- Department of Anatomy & Physiology, Kansas State University, Manhattan, KS 66506
| | - S Ganta
- Department of Anatomy & Physiology, Kansas State University, Manhattan, KS 66506
| | - C Cheng
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506
| | - R Craig
- Department of Anatomy & Physiology, Kansas State University, Manhattan, KS 66506
| | - RR Ganta
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506
| | - LC Freeman
- Department of Anatomy & Physiology, Kansas State University, Manhattan, KS 66506
- *Corresponding author: Lisa C. Freeman, DVM, PhD, Professor of Pharmacology, Kansas State University, 212 Coles Hall, Manhattan, KS 66506-5802, Tel: 785-532-4542, Fax: 785-532-4557,
| |
Collapse
|
77
|
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
|
78
|
Abstract
Epithelial ovarian cancer is a highly fatal disease for which prevention strategies have been limited; in part because of our poor understanding of the underlying biology of its precursor, the ovarian surface epithelium (OSE). The OSE is a single layer of flat-to-cuboidal mesothelial cells that covers the surface of the ovary. Despite its inconspicuous appearance in vivo, it is believed that OSE cells actively participate in the cyclical ovulatory rupture and repair process. The continuous rupture of the OSE at ovulation and the subsequent proliferation to repair the wound renders the cells susceptible to genetic damage and malignant transformation. As the ovary is a rich source of multiple hormones, and normal OSE and ovarian carcinomas secrete and have receptors for hormones, growth factors and cytokines, these factors are strong candidates to regulate normal OSE physiology and the transformation and progression of ovarian cancers. In particular, alterations of hormone/growth factor production and receptor expression are common in ovarian tumors. This review summarizes the current knowledge in the field of endocrinology and its relationship to the biology and pathology of the OSE.
Collapse
Affiliation(s)
- Alice S T Wong
- Department of Zoology, University of Hong Kong, Hong Kong, China.
| | | |
Collapse
|
79
|
Jeoung M, Lee C, Ji I, Ji TH. Trans-activation, cis-activation and signal selection of gonadotropin receptors. Mol Cell Endocrinol 2007; 260-262:137-43. [PMID: 17055146 PMCID: PMC1831837 DOI: 10.1016/j.mce.2005.09.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2005] [Accepted: 09/13/2005] [Indexed: 10/24/2022]
Abstract
It has been thought that when a hormone binds to a receptor, the liganded receptor activates itself and generates hormone signals, such as the cAMP signal and the inositol phosphate signal (cis-activation). We describe that a liganded LH receptor or FSH receptor molecule is capable of intermolecularly activating nonliganded receptors (trans-activation). Particularly, intriguing is the possibility that a pair of compound heterozygous mutants, one defective in binding and the other defective in signaling, may cooperate and rescue signaling. Furthermore, trans-activation of the binding deficient receptors examined in our studies generates either the cAMP signal or the IP signal, but not both. Trans-activation and selective signal generation have broad implications on signal generation mechanisms, and suggest new therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Tae H. Ji
- *Correspondence should be sent to Tae H, Ji, Department of Chemistry, University of Kentucky, Lexington, KY 40506-0055. , Tel: 859-257-3163, Fax:859-257-3229
| |
Collapse
|
80
|
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
|
81
|
Pon YL, Wong AST. Gonadotropin-induced apoptosis in human ovarian surface epithelial cells is associated with cyclooxygenase-2 up-regulation via the beta-catenin/T-cell factor signaling pathway. Mol Endocrinol 2006; 20:3336-50. [PMID: 16945989 DOI: 10.1210/me.2006-0125] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Gonadotropins play a prominent role in ovarian function and pathology. We have shown that treatment with gonadotropins (FSH and LH/human chorionic gonadotropin) reduces the amount of N-cadherin with a concomitant induction of apoptosis in human ovarian surface epithelial (OSE) cells, but precise molecular mechanisms remain to be elucidated. Here, we demonstrated activation of beta-catenin/T-cell factor (TCF) signaling by gonadotropins. We further showed that ectopic expression of N-cadherin was sufficient to recruit beta-catenin to the plasma membrane, thereby blocking beta-catenin/TCF-mediated transactivation in gonadotropin-treated cells. Transfection with beta-catenin small interfering RNA or expression of dominant negative TCF inhibited apoptosis, whereas expression of dominant stable beta-catenin (S37A) caused significant apoptosis, thus supporting a proapoptotic role for beta-catenin/TCF in human OSE. In addition, we showed that gonadotropins enhanced beta-catenin/TCF transcriptional activity through inactivation of glycogen synthase kinase-3beta in a phosphatidylinositol 3-kinase/Akt-dependent manner, indicating cross talk between the phosphatidylinositol 3-kinase/Akt and beta-catenin signaling pathways through glycogen synthase kinase-3beta. Furthermore, gonadotropins increased cyclooxygenase-2 (COX-2) expression via the beta-catenin/TCF pathway. COX-2 also played a role in gonadotropin-induced apoptosis, as treatment with the COX-2-specific inhibitor NS-398 or COX-2 small interfering RNA blocked gonadotropin-dependent apoptotic activity. These findings suggest that the participation of beta-catenin in adhesion and signaling may represent a novel mechanism through which gonadotropins may regulate the cellular fate of human OSE.
Collapse
Affiliation(s)
- Yuen Lam Pon
- Department of Zoology, University of Hong Kong, Pokfulam Road, Hong Kong
| | | |
Collapse
|
82
|
Rzepka-Górska I, Tarnowski B, Chudecka-Głaz A, Górski B, Zielińska D, Tołoczko-Grabarek A. Premature menopause in patients with BRCA1 gene mutation. Breast Cancer Res Treat 2006; 100:59-63. [PMID: 16773440 DOI: 10.1007/s10549-006-9220-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Accepted: 03/02/2006] [Indexed: 12/30/2022]
Abstract
This study was undertaken with regard to the gonadotropin theory of ovarian cancer advocated in the literature and was designed to disclose specific features of ovarian morphology in carriers of the BRCA1 gene mutation. We enrolled 171 patients and divided them into two groups: A (n=90)--operated for breast cancer (30 patients with and 60 without the BRCA1 mutation); B (n=81)--with the BRCA1 mutation qualified for preventive adnexectomy. According to the authors' classification described herein, some patients without the BRCA1 mutation retained "signs of estrogenization" in menopausal ovaries, revealing the role of estrogens as a factor promoting mammary carcinogenesis in these patients. A tendency to premature menopause was observed in BRCA1 mutation carriers of groups A and B as evidenced by the final menorrhea appearing at a younger age and almost total absence of "signs of estrogenization" in menopausal ovaries. It is concluded from these findings that earlier menopause in carriers of the BRCA1 mutation is associated with hypergonadotropic activity and may predispose to ovarian cancer at younger age.
Collapse
Affiliation(s)
- Izabella Rzepka-Górska
- Department of Gynecological Surgery and Oncology of Adults and Adolescents, Pomeranian Medical University of Szczecin, Poland, ul. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland.
| | | | | | | | | | | |
Collapse
|
83
|
Choi JH, Choi KC, Auersperg N, Leung PCK. Gonadotropins activate proteolysis and increase invasion through protein kinase A and phosphatidylinositol 3-kinase pathways in human epithelial ovarian cancer cells. Cancer Res 2006; 66:3912-20. [PMID: 16585220 DOI: 10.1158/0008-5472.can-05-1785] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite evidence that gonadotropins may facilitate peritoneal metastasis of ovarian cancer by increasing cell adhesion, the action and molecular mechanism of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) in ovarian cancer invasion is not well characterized. In the present study, we investigated the effects of FSH and LH on the invasive activity and the expression of metastasis-related proteinases in human epithelial ovarian cancer by Western blot, zymography, reverse transcription-PCR (RT-PCR), ELISA, and Boyden chamber assay. Treatment with FSH or LH (10, 100, or 1,000 ng/mL) significantly increased the invasion of ovarian cancer cell lines, including BG-1, CaOV-3, and SKOV-3 cells but not OVCAR-3 cells. In addition, treatment of SKOV-3 cells with FSH or LH (100 or 1,000 ng/mL) enhanced the expression and activation of matrix metalloproteinases (MMP-2 and MMP-9) as shown by RT-PCR, gelatin zymography, and ELISA. Pretreatment with [(2R)-2-(hydroxamido-carbonylmethyl)-4-methylpentanoyl]-l-tryptophan methylamide (10 micromol/L), a total MMP inhibitor, and 3-(4-phenoxyphenylsulfonyl)-propylthiirane (20 micromol/L), a specific gelatinase inhibitor, neutralized the proinvasive effect of gonadotropins in SKOV-3 cells. In addition, the secretion of tissue inhibitor of metalloproteinases (TIMP-1 and TIMP-2) and plasminogen activator inhibitor-1 was significantly decreased by FSH and LH (100 or 1,000 ng/mL). We further showed that gonadotropins induced an increase in SKOV-3 invasiveness via the activation of protein kinase A (PKA) and phosphatidylinositol 3-kinase (PI3K) signaling pathways. Taken together, these results suggest that gonadotropins may contribute to ovarian cancer metastasis via activation of proteolysis and increase in invasion through the PKA and PI3K pathways.
Collapse
Affiliation(s)
- Jung-Hye Choi
- Department of Obstetrics and Gynecology, British Columbia Children's and Women's Hospital, Child and Family Research Institute, University of British Columbia, 4490 Oak Street, Vancouver, British Columbia, Canada V6H 3V5
| | | | | | | |
Collapse
|
84
|
Konishi I. Gonadotropins and ovarian carcinogenesis: a new era of basic research and its clinical implications. Int J Gynecol Cancer 2006; 16:16-22. [PMID: 16445604 DOI: 10.1111/j.1525-1438.2006.00425.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The possible relationship between gonadotropins and ovarian carcinoma development has received much attention, and in recent years, great progress has been made in basic and epidemiologic research about this issue. Gonadotropins sensitivity in the ovarian surface epithelium (OSE) and in a subset of ovarian carcinomas has been established in vivo and in vitro. Gonadotropins have been shown to induce various biologic actions in the OSE and ovarian carcinoma cells, such as changes in cell proliferation, apoptosis, cell adhesion, and chemosensitivity. These basic studies strongly suggest that gonadotropins are involved in the development and progression of ovarian carcinoma. In contrast, although earlier studies showed a significant risk of infertility therapy for ovarian carcinoma development, subsequent studies reported only slightly increased or no significant increased risk of gonadotropin stimulation and/or assisted reproductive technologies for ovarian carcinoma development. Therefore, the association between ovarian stimulation and ovarian carcinoma remains controversial. Nevertheless, since development of ovarian carcinoma in infertile women during infertility treatment is a serious concern for gynecologists, this review also covers important points for clinical practice, especially the issue of early detection of ovarian carcinoma.
Collapse
Affiliation(s)
- I Konishi
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan.
| |
Collapse
|
85
|
Burdette JE, Kurley SJ, Kilen SM, Mayo KE, Woodruff TK. Gonadotropin-induced superovulation drives ovarian surface epithelia proliferation in CD1 mice. Endocrinology 2006; 147:2338-45. [PMID: 16484319 DOI: 10.1210/en.2005-1629] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ovarian surface epithelium (OSE) is a monolayer of cells that surround the ovary and accommodate repeated tear and repair in response to ovulation. OSE cells are thought to be the progenitors of 90% of ovarian cancers. Currently, the total amount of proliferation of the OSE has not been reported in response to one ovulatory event. In this study, proliferation of the OSE was quantified in response to superovulation induced by ip injection of pregnant mare serum gonadotropin (PMSG) and human chorionic gonadotropin (hCG) in immature 27-d-old CD1 mice using bromodeoxyuridine (BrdU). BrdU incorporation into the OSE cells was measured from the time of hCG injection for a total cumulative label of 12 h. BrdU incorporation was also measured from the time of PMSG injection for a total label of 60 h to correlate proliferation with specific gonadotropin stimulation. The OSE proliferation was significantly higher in superovulated animals compared with control mice at all time points. Proliferation was also analyzed in discrete anatomical sections and indicated that OSE covering antral follicles and corpora lutea proliferated more rapidly than OSE distal to follicular growth. Finally, apoptosis was assessed in response to ovulation, and virtually no cell death within the OSE was detected. These data demonstrate that the OSE, especially near antral follicles and corpora lutea, proliferates significantly in response to the gonadotropins PMSG and hCG. Therefore, ovarian surface cell division in response to ovulation could contribute to ovarian cancer by proliferation-induced DNA mutations and transformed cell progression.
Collapse
Affiliation(s)
- Joanna E Burdette
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208, USA
| | | | | | | | | |
Collapse
|
86
|
Yang CQ, Chan KYK, Ngan HYS, Khoo US, Chiu PM, Chan QKY, Xue WC, Cheung ANY. Single nucleotide polymorphisms of follicle-stimulating hormone receptor are associated with ovarian cancer susceptibility. Carcinogenesis 2006; 27:1502-6. [PMID: 16574671 DOI: 10.1093/carcin/bgl014] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Epidemiological studies suggested that ovulation was associated with ovarian carcinogenesis. Follicle-stimulating hormone (FSH) played an important role in follicular development and was recently found to affect growth of ovarian epithelial cells. Single nucleotide polymorphisms (SNPs) Thr307Ala and Asn680Ser were two non-synonymous variations in the coding region of the FSH receptor (FSHR) gene. This hitherto first case-control study investigating the association between these two FSHR SNPs and the risk of ovarian cancer involved 202 histopathologically confirmed ovarian cancer patients and 266 age-matched cancer-free control subjects using restriction fragment length polymorphism assay and direct sequencing. Our results demonstrated that the 307Ala and 680Ser carriers were associated with significantly increased risk of developing serous and mucinous types of ovarian cancers (P < 0.0005, OR = 2.60, 95% CI = 1.56-4.34; and P < 0.0005, OR = 2.89, 95% CI = 1.73-4.84, adjusted for age, respectively) but not endometrioid and clear cell types. The two SNPs were found to be in modest linkage disequilibrium, D' = 0.804 and 0.701, r2 = 0.581 and 0.406 for the cancer and control groups, respectively. The major haplotype of 307Ala-680Ser was also associated with higher cancer risk (P = 0.033, OR = 1.39, 95% CI = 1.03-1.88), especially for the serous and mucinous carcinomas (P = 0.001, OR = 1.82, 95% CI = 1.27-2.60). Our results suggested that the two FSHR SNPs might affect the susceptibility of women to specific subtypes of ovarian cancer. Different types of ovarian cancer might adopt distinct carcinogenetic pathways. Such understanding may be important in selecting patients for ovulation induction therapy.
Collapse
Affiliation(s)
- C Q Yang
- Department of Pathology, Jockey Club Clinical Research Centre, The University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Edmondson RJ, Monaghan JM, Davies BR. Gonadotropins mediate DNA synthesis and protection from spontaneous cell death in human ovarian surface epithelium. Int J Gynecol Cancer 2006; 16:171-7. [PMID: 16445629 DOI: 10.1111/j.1525-1438.2006.00274.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Gonadotropins have been implicated in the development of epithelial ovarian cancers. These tumors are derived from ovarian surface epithelium (OSE). The purpose of this study was to determine the effects of these hormones on DNA synthesis and spontaneous cell death in primary cultures of OSE and three immortalized OSE cultures. Primary cultures of OSE cells were generated from the ovaries of women with benign disease. The effects of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) on DNA synthesis and cell death were determined using [(3)H]thymidine incorporation and JAM assays. Significant inductions of DNA synthesis were demonstrated with LH in 4/12 (33%) primary cultures of OSE and 2/3 OSE cell lines and with FSH in 4/11 (36%) primary cultures of OSE and 2/3 OSE cell lines. A significant protection from cell death was also observed in the presence of FSH in 2/4 primary cultures of OSE and 1/3 OSE cell lines and in the presence of LH in 1/4 primary cultures of OSE and 2/3 OSE cell lines. The results indicate that while gonadotropins have the potential to induce cell proliferation and protect from cell death in OSE cells in vitro, their effects are variable in OSE cells from different women.
Collapse
Affiliation(s)
- R J Edmondson
- Northern Gynaecological Oncology Centre, Queen Elizabeth Hospital, Gateshead, Tyne and Wear NE9 6SX, UK.
| | | | | |
Collapse
|
88
|
Abstract
Two much-debated hypotheses regarding the aetiology of epithelial ovarian cancer (EOC) are incessant ovulation and gonadotrophin stimulation. A significant inverse correlation is found between the risk of EOC and number of pregnancies, duration of oral contraceptive use (about 10% risk reduction per year) and duration of breastfeeding. Whereas ovulation in the 20-29 year age group was associated with a 20% increase in risk for each year of ovulation, maximum protective effect was noticed in late childbirth (>35 years). Although both in-vitro and in-vivo studies showed that gonadotrophins may initiate and stimulate the growth of EOC, it is not known whether gonadotrophins promote EOC. FSH and LH receptors have been detected by ligand-binding assay and by reverse transcriptase polymerase chain reaction in ovarian surface epithelium (OSE) and in malignant epithelial ovarian tissue. Key aetiological events for this cancer may occur in the premenopausal period. Although hormone replacement therapy (HRT) can lower FSH and LH concentrations substantially, it cannot reduce EOC. The ideal time of ovulation and/or gonadotrophin suppression for prevention of EOC, and why oral contraceptives (OC) can but HRT cannot reduce EOC effectively, are unknown. Various growth-related genes, factors, adhesion molecules and angiogenic factors are present in OSE. Pituitary ovarian axis hormones seem to maintain a delicate balance towards growth control whilst the resultant chain of abnormal growth-promoting events occur at cellular level. Reports regarding the relation of exogenous and endogenous hormones, especially oestrogen, and post-menopausal EOC are reviewed.
Collapse
Affiliation(s)
- Chinmoy K Bose
- Health Department, Kolkata Municipal Corporation, Kolkata 700 013, India.
| |
Collapse
|
89
|
Ahtiainen P, Rulli SB, Shariatmadari R, Pelliniemi LJ, Toppari J, Poutanen M, Huhtaniemi IT. Fetal but not adult Leydig cells are susceptible to adenoma formation in response to persistently high hCG level: a study on hCG overexpressing transgenic mice. Oncogene 2005; 24:7301-9. [PMID: 16007123 DOI: 10.1038/sj.onc.1208893] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We have previously demonstrated that male transgenic (TG) mice overexpressing human chorionic gonadotropin (hCG+) develop reproductive organ defects, but no tumors, in adult age. In this study, the effects of persistently elevated hCG were followed in TG males between day 5 postpartum and adulthood. Leydig cell (LC) adenomas were found in prepubertal mice, most prominently at the age of 10 days, but not in adult age. Serum testosterone concentrations were significantly increased in TG males at all ages studied. The phenotype of the prepubertal hCG+ males resembled that found in boys upon expression of constitutively activating luteinizing hormone (LH) receptor mutations. The temporal expression patterns of the fetal LC marker gene, thrombospondin 2, and those of adult LCs, hydroxysteroid dehydrogenase-6, delta5-3-beta and prostaglandin D synthase, were similar in wild-type and hCG+ males. Hence, the postnatal adenomas resemble functionally fetal LCs, and only these cells are susceptible to hCG-induced tumorigenesis. Our findings demonstrate a novel intriguing difference between the fetal and adult LC populations and provide further insight into the potential tumorigenic effects of gonadotropins.
Collapse
Affiliation(s)
- Petteri Ahtiainen
- Department of Physiology, University of Turku, FIN-20520 Turku, Finland
| | | | | | | | | | | | | |
Collapse
|
90
|
Pasapera AM, Jiménez-Aguilera MDP, Chauchereau A, Milgrom E, Olivares A, Uribe A, Gutiérrez-Sagal R, Ulloa-Aguirre A. Effects of FSH and 17beta-estradiol on the transactivation of estrogen-regulated promoters and cell proliferation in L cells. J Steroid Biochem Mol Biol 2005; 94:289-302. [PMID: 15857748 DOI: 10.1016/j.jsbmb.2004.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Accepted: 11/29/2004] [Indexed: 11/26/2022]
Abstract
In the present study, we analyzed human follicle-stimulating hormone (FSH)-induced cell proliferation and transactivation of estrogen-sensitive reporter genes-in L cells stably expressing the human FSH receptor [L-(hFSHR(+)) cells]. In order to dissect the signaling pathways involved in this process, L-(hFSHR(+)) cells were transiently transfected with either the 3X-ERE-TAT-Luc or the ERE-VitA2-TK-CAT reporter genes and treated with FSH or PKA activators (cholera toxin, forskolin and 8-Br-cAMP) in the presence or absence of various kinase inhibitors. We found that FSH and all PKA activators, specifically induced transactivation of both reporter genes. Transactivation of estrogen-sensitive genes by FSH or PKA activators were blocked (approximately 90%) by H89 (PKA inhibitor) and LY294002 but not by Wortmannin (PI3-K inhibitors), 4-OH-tamoxifen, ICI182,780 or SB203580 (p38 MAPK inhibitor); PD98059 (ERK1/2 inhibitor) partially (approximately 30%) blocked the FSH-mediated effect. The combination of FSH and estradiol resulted in a synergistic effect on transactivation as well as on cell proliferation, and this enhancement was attenuated by antiestrogens. We additionally analyzed the participation of the coactivators SRC-1 and cAMP response element binding protein (CREB)-binding protein (CBP) in FSH-evoked estrogen receptor (ER)-dependent transactivation; we found that CBP but not SRC-1 potentiated FSH-induced transcriptional activation of both ER-sensitive reporters, being this effect stronger on the ERE-VitA2-TK-CAT than on the 3X-ERE-TAT-Luc reporter. Thus, in L-(hFSHR(+)) cells FSH induces transcriptional activation of estrogen-sensitive genes through an A-kinase-triggered signaling pathway, using also to a lesser extent the ERK1/2 and p38 pathways. PI3-K is not apparently involved in this FSH-mediated process since LY294002, but not Wortmannin, specifically binds ERs and completely blocks estrogen action. Presumably, CBP cooperates with the ER on genes that contain estrogen responsive elements through mechanisms involving the participation of other proteins and/or basal transcription factors (e.g. CREB), which in turn mediate the transcriptional response of estrogen-sensitive reporter genes to FSH stimulation.
Collapse
Affiliation(s)
- Ana María Pasapera
- Research Unit in Reproductive Medicine, Hospital de Gineco-Obstetricia Luis Castelazo Ayala, Instituto Mexicano del Seguro Social, Apdo. Postal 99-065, Unidad Independencia, México D.F. C.P. 10101, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Pon YL, Auersperg N, Wong AST. Gonadotropins regulate N-cadherin-mediated human ovarian surface epithelial cell survival at both post-translational and transcriptional levels through a cyclic AMP/protein kinase A pathway. J Biol Chem 2005; 280:15438-48. [PMID: 15701645 DOI: 10.1074/jbc.m410766200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gonadotropins are the major regulators of ovarian function and may be involved in the etiology of ovarian cancer. In this study, we report a new mechanism whereby gonadotropins regulate the survival of human ovarian surface epithelium (OSE), the tissue of origin of epithelial ovarian carcinomas. Our results indicate that disruption of N-cadherin-mediated cell-cell adhesion is an important molecular event in the apoptosis of human OSE. Treatment with surge serum concentrations of gonadotropins reduced the amount of N-cadherin with a concomitant induction of apoptosis, and this effect was mediated by a cAMP/protein kinase A pathway but not the ERK1/2 and protein kinase C cascades. We further demonstrated that activation of the gonadotropins/cAMP signaling pathway in human OSE led to a rapid down-regulation of N-cadherin protein level followed by a reduction at the level of N-cadherin mRNA, indicating that expression of N-cadherin was regulated by post-translational and transcriptional mechanisms. The former mechanism was mediated by increased turnover of N-cadherin protein and could be reversed by inhibition of proteasomal or matrix metalloproteinase (MMP-2) activity. On the other hand, at the transcriptional level, the addition of actinomycin D abolished the cAMP-mediated decrease in N-cadherin mRNA but did not change its stability. Inhibition of protein kinase A or expressing a dominant negative mutant of cAMP-response element-binding protein blocked this decrease of N-cadherin mRNA. Together, the combined operation of post-translational and transcriptional mechanisms suggests that regulation of N-cadherin is a crucial event and emphasizes the important role that N-cadherin has in controlling the survival capability of human OSE.
Collapse
Affiliation(s)
- Yuen Lam Pon
- Department of Zoology, University of Hong Kong, Hong Kong
| | | | | |
Collapse
|
92
|
Abstract
The neurotrophins (NT) including nerve growth factor (NGF) are a family of related growth factors that are of major importance in the regulation of neuronal survival and differentiation. In the ovary, they can help in follicular maturation and ovulation by inducing the FSH receptor (FSHR). Current literature shows that perimenopausal ovarian surface epithelium (OSE) can also express FSHR. By G protein link, this FSHR is capable of precipitating neoplasia of OSE, which is the commonest in the ovary. NT are implicated as the cause of this aberrant expression of FSHR in OSE. By central action NT can lower serum FSH, as is found in ovarian cancer. Thus, NGF deregulates expression of FSHR in OSE and secretion of FSH from the pituitary. This phenomenon may hold the key to the hitherto unexplained carcinogenic process of sporadic epithelial ovarian cancer.
Collapse
|
93
|
Gubbay O, Guo W, Rae MT, Niven D, Howie AF, McNeilly AS, Xu L, Hillier SG. Anti-inflammatory and proliferative responses in human and ovine ovarian surface epithelial cells. Reproduction 2004; 128:607-14. [PMID: 15509706 DOI: 10.1530/rep.1.00272] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The majority of ovarian cancers (>90%) are believed to derive from the ovarian surface epithelium (OSE); a single layer covering the entire surface of the ovary. At ovulation, the OSE cell layer undergoes an inflammatory response, involving cell death and growth, in order to overcome ovarian surface rupture. Abnormalities during these processes are believed to contribute to the development of tumours. Using primary cultures of OSE cells, we have compared anti-inflammatory and proliferative responses directly between human and ovine OSE cells to further establish the use of ovine OSE cells as a suitable model system for the study of human OSE cells. In order to compare effects of inflammatory stimulation, expression and activity of 11βhydroxysteroid dehydrogenase (11βHSD) type 1 was measured in OSE cells in response to interleukin (IL)-1α. As previously identified in human OSE cells, treatment of ovine OSE cells with IL-1α stimulated a concomitant increase of 11βHSD type 1 mRNA (31-fold;P< 0.05) and oxoreductase activity, indicating an increased production of anti-inflammatory cortisol. To compare the growth of human and ovine OSE cells, OSE cell number was measured in response to treatment with gonadotropins or growth factors. In the presence of FSH, LH or human chorionic gonadotropin (hCG), ovine and human OSE cell growth was similarly stimulated >1.2-fold (P< 0.05). In the presence of connective tissue growth factor (CTGF) and more significantly insulin growth factor I (IGF-I), human and ovine OSE cell growth was also similarly stimulated >1.2-fold (P< 0.05) and >1.5-fold (P< 0.01), respectively. The induction of both human and ovine OSE cell growth by IGF-I or hCG was further shown to be dependent on activation of the MAP kinase/extracellular-signal-regulated kinase (ERK) pathway. Stimulation of ovine OSE cell growth by hepatocyte growth factor (HGF) was similarly shown to be ERK-dependent; however, for human OSE cells, HGF only mildly stimulated ERK phosphorylation and failed to stimulate OSE cell growth. The demonstration that human and ovine OSE cells share similarities at the level of cell signalling, gene expression and cellular growth supports the use of ovine OSE cells as a suitable model for the study of human OSE cells.
Collapse
Affiliation(s)
- O Gubbay
- Department of Reproductive and Developmental Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Old Dalkeith Road, Edinburgh EH16 4SB, UK.
| | | | | | | | | | | | | | | |
Collapse
|
94
|
Gadducci A, Cosio S, Gargini A, Genazzani AR. Sex-steroid hormones, gonadotropin and ovarian carcinogenesis: a review of epidemiological and experimental data. Gynecol Endocrinol 2004; 19:216-28. [PMID: 15724805 DOI: 10.1080/09513590400014354] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- A Gadducci
- Department of Procreative Medicine and Child Development, Division of Gynecology and Obstetrics, University of Pisa, Italy
| | | | | | | |
Collapse
|
95
|
Piek JMJ, Kenemans P, Verheijen RHM. Intraperitoneal serous adenocarcinoma: a critical appraisal of three hypotheses on its cause. Am J Obstet Gynecol 2004; 191:718-32. [PMID: 15467531 DOI: 10.1016/j.ajog.2004.02.067] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Serous ovarian, Fallopian tube, and peritoneal adenocarcinomas are remarkably similar, both in their morphology, as well as in their clinical behavior. Despite extensive clinical and fundamental research, controversy still exists on the origin of serous female adnexal tumors. Difficulties in identification of site of origin at late stage the of disease at detection, when ovary, Fallopian tube, and the abdominal cavity are usually all involved, in addition to their macroscopic and microscopic resemblance, are major causes of this debate. In 3 hypotheses, 3 possible tissues of origin are proposed: the ovarian surface epithelium, the Fallopian tube epithelium, and the secondary Mullerian system. STUDY DESIGN We searched for all peer-reviewed articles and reviews that examined "serous ovarian carcinoma," "Fallopian tube carcinoma," "Mullerian system," "ovarian surface epithelium," "tubal epithelium," and "peritoneal." We included only articles that could give information on the origin of serous carcinomas. Additional articles were added by examining references of overview articles in relevant fields. RESULTS Discussed are the experimental data underlying these hypotheses. CONCLUSION An attempt is made to integrate the 3 hypotheses into a comprehensive model of serous intraperitoneal adenocarcinogenesis. It can be concluded that the Fallopian tubes play a major role in the development of female serous cancer.
Collapse
Affiliation(s)
- Jurgen M J Piek
- Department of Obstetrics and Gynecology, Vrije University Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
96
|
Steinmetz R, Wagoner HA, Zeng P, Hammond JR, Hannon TS, Meyers JL, Pescovitz OH. Mechanisms regulating the constitutive activation of the extracellular signal-regulated kinase (ERK) signaling pathway in ovarian cancer and the effect of ribonucleic acid interference for ERK1/2 on cancer cell proliferation. Mol Endocrinol 2004; 18:2570-82. [PMID: 15243131 DOI: 10.1210/me.2004-0082] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The ERK1/2 MAPK pathway is a critical signaling system that mediates ligand-stimulated signals for the induction of cell proliferation, differentiation, and cell survival. Studies have shown that this pathway is constitutively active in several human malignancies and may be involved in the pathogenesis of these tumors. In the present study we examined the ERK1/2 pathway in cell lines derived from epithelial and granulosa cell tumors, two distinct forms of ovarian cancer. We show that ERK1 and ERK2 are constitutively active and that this activation results from both MAPK kinase-dependent and independent mechanisms and is correlated with elevated BRAF expression. MAPK phosphatase 1 (MKP-1) expression, which is involved in ERK1/2 deactivation, is down-regulated in the cancer cells, thus further contributing to ERK hyperactivity in these cells. Treatment of these cancer cell lines with the proteasome inhibitor ZLLF-CHO increased MKP-1 but not MKP-2 expression and decreased ERK1/2 phosphorylation. More importantly, silencing of ERK1/2 protein expression using RNA interference led to the complete suppression of tumor cell proliferation. These results provide evidence that the ERK pathway plays a major role in ovarian cancer pathogenesis and that down-regulation of this master signaling pathway is highly effective for the inhibition of ovarian tumor growth.
Collapse
Affiliation(s)
- Rosemary Steinmetz
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, James Whitcomb Riley Hospital for Children, Indianapolis, Indiana 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
97
|
Funaro A, Sapino A, Ferranti B, Horenstein AL, Castellano I, Bagni B, Garotta G, Malavasi F. Functional, structural, and distribution analysis of the chorionic gonadotropin receptor using murine monoclonal antibodies. J Clin Endocrinol Metab 2003; 88:5537-46. [PMID: 14602802 DOI: 10.1210/jc.2003-030977] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
LH and human chorionic gonadotropin (hCG) control steroid production and gametogenesis. They also function as growth factors through interaction with a specific receptor that is a member of the seven-transmembrane receptor family coupled via G proteins to signal pathways involving cAMP and phospholipase C/inositol 3 phosphate. For this study, monoclonal antibodies (mAbs) were raised against the human LH receptor (LHR)/hCG receptor (hCGR), using Chinese hamster ovary LHR-transfected cells as the immunogen. Two reagents were then selected on the basis of their ability to recognize the full-length transmembrane receptor expressed both by Chinese hamster ovary LHR-transfected cells and by a limited number of tumor cell lines. One of these mAbs reacts with the LHR/hCGR in tissue sections of both frozen and paraffin-embedded specimens. This unique feature allowed us to map the cytological distribution of LHR/hCGR in human breast tissues at different stages of development in physiological and benign pathological conditions. The same mAb proved to be agonistic: receptor ligation elicits signals that modulate the growth of selected breast tumor cell lines. This observation suggests that the mAb recognizes an epitope that is included in the domain of the receptor involved in the interaction with the natural ligand.
Collapse
Affiliation(s)
- Ada Funaro
- Department of Genetics, Biology and Biochemistry and Research Center for Experimental Medicine, University of Torino, Torino 10126, Italy
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Ho SM, Lau KM, Mok SCH, Syed V. Profiling follicle stimulating hormone-induced gene expression changes in normal and malignant human ovarian surface epithelial cells. Oncogene 2003; 22:4243-56. [PMID: 12833147 DOI: 10.1038/sj.onc.1206437] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Epidemiological data have implicated the pituitary gonadotropin follicle stimulating hormone (FSH) as both a risk factor for and a protective agent against epithelial ovarian cancer. Yet, little is known about how this hormone could play such opposing roles in ovarian carcinogenesis. Complementary DNA microarrays containing 2400 named genes were used to examine FSH-induced gene expression changes in ovarian cancer (OC) and immortalized normal human ovarian surface epithelial (HOSE) cell lines. Two-way t-statistics analyses of array data identified two distinct sets of FSH-regulated genes in HOSE and in established OC cell lines established from patients (OVCA cell lines). Among the HOSE cell lines, FSH increased expression of 57% of the 312 genes and downregulated 43%. In contrast, FSH diminished expression of 92% of the 177 genes in the OVCA cell lines. All but 18 of the genes affected by FSH in HOSE cell lines were different from those altered in OVCA cell lines. Among the 18 overlapping genes, nine genes exhibited the same direction of change following FSH challenge, while the other nine showed discordance in response between HOSE and OVCA cell lines. The FSH-induced differential expression of seven out of nine genes was confirmed by real-time RT-PCR. Gene-specific antisense oligonuleotides (ODNs) were used to inhibit the expression of genes encoding GTPase activating protein (rap1GAP), neogenin, and restin in HOSE and OVCA cells. Antisense ODNs to neogenin and restin, but not an antisense ODN to rap1GAP, were effective in inhibiting OVCA cell growth, diminishing proliferating cell nuclear antigen expression, and increasing caspase 3 activities. Furthermore, the ODN to rap1GAP was further shown to be ineffective in altering migration properties of OVCA cell lines. HOSE cell proliferation was not affected by treatment with any of the antisense ODNs. In summary, gene profiling data reveal for the first time that FSH may exert different biological actions on OVCA cells than on HOSE cells, by differential regulation of a set of putative oncogenes/tumor suppressors. Specifically, neogenin and restin were found to exhibit proproliferation/survival action on OC cells.
Collapse
Affiliation(s)
- Shuk-Mei Ho
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
99
|
Wang J, Lin L, Parkash V, Schwartz PE, Lauchlan SC, Zheng W. Quantitative analysis of follicle-stimulating hormone receptor in ovarian epithelial tumors: a novel approach to explain the field effect of ovarian cancer development in secondary mullerian systems. Int J Cancer 2003; 103:328-34. [PMID: 12471615 DOI: 10.1002/ijc.10848] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The role of FSHR expression in ovarian cancer development is not clear. We examined quantitative expression of FSHR in different types of OET, presumed precursor lesions and peritoneal implants and further discussed FSH as a key growth-promotion factor for the process of ovarian epithelial tumorigenesis. Thirty-five primary OET specimens, including 5 serous cystadenomas, 4 papillary serous cystadenomas, 9 SBTs and 17 serous carcinomas, were examined for quantitative FSHR expression. Ten paired samples (3 benign cystadenomas, 5 SBTs and 2 carcinomas) were obtained from several morphologically different areas, including benign-looking, borderline and cancerous areas in the same OETs, and from the remaining ovarian tissue and contralateral ovaries. Competitive RT-PCR was performed to measure the quantitative expression of FSHR in each tissue sample. FSHR expression levels were compared among nonpaired samples and within paired samples. We found that OSE had the lowest FSHR expression, whereas antral follicles had the highest level. Within benign OETs, papillary serous cystadenomas have 4.9-fold higher FSHR levels than nonpapillary serous cystadenomas. SBTs had the highest level of FSHR expression, which was 12.8-fold, 2.7-fold and 2.4-fold higher than that of serous cystadenomas, papillary serous cystadenomas and grade 1 carcinomas, respectively. A similarly high level of FSHR mRNA was found in peritoneal implants, which were associated with SBTs. FSHR levels among serous carcinomas decreased with an increase in carcinoma grade. Grade 3 carcinomas had the lowest FSHR level, which was similar to that of serous cystadenomas, while grade 1 carcinomas had 6.5-fold higher FSHR levels than those in serous cystadenomas. Our results suggest that not only serum FSH but also FSHR in ovarian epithelium may play important roles in ovarian OET development. Both the receptor and ligand may act in a synergistic way to promote tumor growth. The observation that high FSHR levels are present in peritoneal implants suggests that FSH may also play a similar role in the development of peritoneal serous tumors. From this perspective, circulating FSH may be considered a driving force in the field effect theory for the development of both ovarian neoplasms and their associated peritoneal implants. However, the exact role of FSH and/or FSHR in the development of epithelial tumors arising in both the ovary and peritoneum needs further investigation.
Collapse
MESH Headings
- Adult
- Aged
- Case-Control Studies
- Cystadenoma/genetics
- Cystadenoma/metabolism
- Cystadenoma/pathology
- Cystadenoma, Papillary/genetics
- Cystadenoma, Papillary/metabolism
- Cystadenoma, Papillary/pathology
- Cystadenoma, Serous/genetics
- Cystadenoma, Serous/metabolism
- Cystadenoma, Serous/pathology
- DNA Primers/chemistry
- Epithelium/metabolism
- Female
- Humans
- Middle Aged
- Mullerian Ducts/pathology
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Ovary/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, FSH/genetics
- Receptors, FSH/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Jun Wang
- Department of Preventive Medicine, University of Southern California, Los Angeles, USA
| | | | | | | | | | | |
Collapse
|
100
|
Auersperg N. Specific keynote: experimental models of epithelial ovarian carcinogenesis. Gynecol Oncol 2003; 88:S47-51; discussion S52-5. [PMID: 12586085 DOI: 10.1006/gyno.2002.6683] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|