Skip to main content
Top
Published in: Discover Oncology 4/2010

01-08-2010

Expression of Membrane Progesterone Receptors (mPR/PAQR) in Ovarian Cancer Cells: Implications for Progesterone-Induced Signaling Events

Authors: Nathan J. Charles, Peter Thomas, Carol A. Lange

Published in: Discover Oncology | Issue 4/2010

Login to get access

Abstract

The high mortality rates associated with ovarian cancer are largely due to a lack of highly effective treatment options for advanced stage disease; a time when initial diagnosis most commonly occurs. Recent evidence suggests that the steroid hormone, progesterone, may possess anti-tumorigenic properties. With the discovery of a new class of membrane-bound progesterone receptors (mPRs) belonging to the progestin and adipoQ receptor (PAQR) gene family in the ovary, there are undefined mechanisms by which progesterone may inhibit tumor progression. Therefore, our goal was to define potential mPR-dependent signaling mechanisms operative in ovarian cancer cells. We detected abundant mPRα (PAQR7), mPRβ (PAQR8), and mPRγ (PAQR5), but not classical nuclear PR (A or B isoforms) mRNA expression and mPRα protein expression in a panel of commonly used ovarian cancer cell lines. In contrast to mPR action in breast cancer cells, progesterone alone failed to induce changes in cyclic adenosine monophosphate (cAMP) levels in ovarian cancer cells. However, progesterone enhanced cAMP production by β1,2-adrenergic receptors and increased isoproterenol-induced transcription from a cAMP response element (CRE)-driven reporter gene. Independently of β-adrenergic signaling, we additionally observed activation of both JNK1/2 and p38 MAPK in response to progesterone alone. This finding was supported by the results of a screen for potential mPR gene targets. Progesterone induced a significant increase in transcription of the pro-apoptotic marker BAX, whose activity and expression has been linked to JNK1/2 and p38 signaling. Inhibitors of JNK, but not p38, blocked progesterone-induced BAX expression. Taken together, these observations implicate at least two distinct signaling pathways that may be utilized by mPRs in ovarian cancer cells that exhibit regulatory genomic changes. These studies on mPR signaling in ovarian cancer lay the foundation for future work aimed at understanding how progesterone exerts its anti-tumorigenic effects in the ovary and suggest that pharmacologic activation of mPRs, abundantly expressed in ovarian cancers, may provide a new treatment option for patients with advanced stage disease.
Literature
1.
go back to reference Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ (2009) Cancer statistics, 2009. CA Cancer J Clin 59:225–249CrossRefPubMed Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ (2009) Cancer statistics, 2009. CA Cancer J Clin 59:225–249CrossRefPubMed
3.
go back to reference Leung PC, Choi JH (2007) Endocrine signaling in ovarian surface epithelium and cancer. Hum Reprod Update 13:143–162CrossRefPubMed Leung PC, Choi JH (2007) Endocrine signaling in ovarian surface epithelium and cancer. Hum Reprod Update 13:143–162CrossRefPubMed
4.
go back to reference Adami HO, Hsieh CC, Lambe M, Trichopoulos D, Leon D, Persson I et al (1994) Parity, age at first childbirth, and risk of ovarian cancer. Lancet 344:1250–1254CrossRefPubMed Adami HO, Hsieh CC, Lambe M, Trichopoulos D, Leon D, Persson I et al (1994) Parity, age at first childbirth, and risk of ovarian cancer. Lancet 344:1250–1254CrossRefPubMed
5.
go back to reference Thomas HV, Murphy MF, Key TJ, Fentiman IS, Allen DS, Kinlen LJ (1998) Pregnancy and menstrual hormone levels in mothers of twins compared to mothers of singletons. Ann Hum Biol 25:69–75CrossRefPubMed Thomas HV, Murphy MF, Key TJ, Fentiman IS, Allen DS, Kinlen LJ (1998) Pregnancy and menstrual hormone levels in mothers of twins compared to mothers of singletons. Ann Hum Biol 25:69–75CrossRefPubMed
6.
go back to reference Salazar-Martinez E, Lazcano-Ponce EC, Gonzalez Lira-Lira G, Escudero-De los Rios P, Salmeron-Castro J, Hernandez-Avila M (1999) Reproductive factors of ovarian and endometrial cancer risk in a high fertility population in Mexico. Cancer Res 59:3658–3662PubMed Salazar-Martinez E, Lazcano-Ponce EC, Gonzalez Lira-Lira G, Escudero-De los Rios P, Salmeron-Castro J, Hernandez-Avila M (1999) Reproductive factors of ovarian and endometrial cancer risk in a high fertility population in Mexico. Cancer Res 59:3658–3662PubMed
7.
go back to reference Tambyraja AL, Sengupta F, MacGregor AB, Bartolo DC, Fearon KC (2004) Patterns and clinical outcomes associated with routine intravenous sodium and fluid administration after colorectal resection. World J Surg 28:1046–1051, discussion 1051–1042CrossRefPubMed Tambyraja AL, Sengupta F, MacGregor AB, Bartolo DC, Fearon KC (2004) Patterns and clinical outcomes associated with routine intravenous sodium and fluid administration after colorectal resection. World J Surg 28:1046–1051, discussion 1051–1042CrossRefPubMed
8.
go back to reference Modan B, Ron E, Lerner-Geva L, Blumstein T, Menczer J, Rabinovici J et al (1998) Cancer incidence in a cohort of infertile women. Am J Epidemiol 147:1038–1042PubMed Modan B, Ron E, Lerner-Geva L, Blumstein T, Menczer J, Rabinovici J et al (1998) Cancer incidence in a cohort of infertile women. Am J Epidemiol 147:1038–1042PubMed
9.
go back to reference Syed V, Ulinski G, Mok SC, Yiu GK, Ho SM (2001) Expression of gonadotropin receptor and growth responses to key reproductive hormones in normal and malignant human ovarian surface epithelial cells. Cancer Res 61:6768–6776PubMed Syed V, Ulinski G, Mok SC, Yiu GK, Ho SM (2001) Expression of gonadotropin receptor and growth responses to key reproductive hormones in normal and malignant human ovarian surface epithelial cells. Cancer Res 61:6768–6776PubMed
10.
go back to reference Gross TP, Schlesselman JJ, Stadel BV, Yu W, Lee NC (1992) The risk of epithelial ovarian cancer in short-term users of oral contraceptives. Am J Epidemiol 136:46–53PubMed Gross TP, Schlesselman JJ, Stadel BV, Yu W, Lee NC (1992) The risk of epithelial ovarian cancer in short-term users of oral contraceptives. Am J Epidemiol 136:46–53PubMed
11.
go back to reference Rosenberg L, Palmer JR, Zauber AG, Warshauer ME, Lewis JL Jr, Strom BL et al (1994) A case-control study of oral contraceptive use and invasive epithelial ovarian cancer. Am J Epidemiol 139:654–661PubMed Rosenberg L, Palmer JR, Zauber AG, Warshauer ME, Lewis JL Jr, Strom BL et al (1994) A case-control study of oral contraceptive use and invasive epithelial ovarian cancer. Am J Epidemiol 139:654–661PubMed
12.
go back to reference Ness RB, Grisso JA, Vergona R, Klapper J, Morgan M, Wheeler JE (2001) Oral contraceptives, other methods of contraception, and risk reduction for ovarian cancer. Epidemiology 12:307–312CrossRefPubMed Ness RB, Grisso JA, Vergona R, Klapper J, Morgan M, Wheeler JE (2001) Oral contraceptives, other methods of contraception, and risk reduction for ovarian cancer. Epidemiology 12:307–312CrossRefPubMed
13.
go back to reference Greer JB, Modugno F, Allen GO, Ness RB (2005) Short-term oral contraceptive use and the risk of epithelial ovarian cancer. Am J Epidemiol 162:66–72CrossRefPubMed Greer JB, Modugno F, Allen GO, Ness RB (2005) Short-term oral contraceptive use and the risk of epithelial ovarian cancer. Am J Epidemiol 162:66–72CrossRefPubMed
14.
go back to reference Beral V, Doll R, Hermon C, Peto R, Reeves G (2008) Ovarian cancer and oral contraceptives: collaborative reanalysis of data from 45 epidemiological studies including 23, 257 women with ovarian cancer and 87, 303 controls. Lancet 371:303–314CrossRefPubMed Beral V, Doll R, Hermon C, Peto R, Reeves G (2008) Ovarian cancer and oral contraceptives: collaborative reanalysis of data from 45 epidemiological studies including 23, 257 women with ovarian cancer and 87, 303 controls. Lancet 371:303–314CrossRefPubMed
15.
go back to reference Rodriguez GC, Walmer DK, Cline M, Krigman H, Lessey BA, Whitaker RS et al (1998) Effect of progestin on the ovarian epithelium of macaques: cancer prevention through apoptosis? J Soc Gynecol Investig 5:271–276CrossRefPubMed Rodriguez GC, Walmer DK, Cline M, Krigman H, Lessey BA, Whitaker RS et al (1998) Effect of progestin on the ovarian epithelium of macaques: cancer prevention through apoptosis? J Soc Gynecol Investig 5:271–276CrossRefPubMed
16.
go back to reference Rodriguez GC, Nagarsheth NP, Lee KL, Bentley RC, Walmer DK, Cline M et al (2002) Progestin-induced apoptosis in the Macaque ovarian epithelium: differential regulation of transforming growth factor-beta. J Natl Cancer Inst 94:50–60PubMed Rodriguez GC, Nagarsheth NP, Lee KL, Bentley RC, Walmer DK, Cline M et al (2002) Progestin-induced apoptosis in the Macaque ovarian epithelium: differential regulation of transforming growth factor-beta. J Natl Cancer Inst 94:50–60PubMed
17.
go back to reference Lee BH, Hecht JL, Pinkus JL, Pinkus GS (2002) WT1, estrogen receptor, and progesterone receptor as markers for breast or ovarian primary sites in metastatic adenocarcinoma to body fluids. Am J Clin Pathol 117:745–750CrossRefPubMed Lee BH, Hecht JL, Pinkus JL, Pinkus GS (2002) WT1, estrogen receptor, and progesterone receptor as markers for breast or ovarian primary sites in metastatic adenocarcinoma to body fluids. Am J Clin Pathol 117:745–750CrossRefPubMed
18.
go back to reference Noguchi T, Kitawaki J, Tamura T, Kim T, Kanno H, Yamamoto T et al (1993) Relationship between aromatase activity and steroid receptor levels in ovarian tumors from postmenopausal women. J Steroid Biochem Mol Biol 44:657–660CrossRefPubMed Noguchi T, Kitawaki J, Tamura T, Kim T, Kanno H, Yamamoto T et al (1993) Relationship between aromatase activity and steroid receptor levels in ovarian tumors from postmenopausal women. J Steroid Biochem Mol Biol 44:657–660CrossRefPubMed
19.
go back to reference Akahira J, Suzuki T, Ito K, Kaneko C, Darnel AD, Moriya T et al (2002) Differential expression of progesterone receptor isoforms A and B in the normal ovary, and in benign, borderline, and malignant ovarian tumors. Jpn J Cancer Res 93:807–815PubMed Akahira J, Suzuki T, Ito K, Kaneko C, Darnel AD, Moriya T et al (2002) Differential expression of progesterone receptor isoforms A and B in the normal ovary, and in benign, borderline, and malignant ovarian tumors. Jpn J Cancer Res 93:807–815PubMed
20.
go back to reference Lau KM, Mok SC, Ho SM (1999) Expression of human estrogen receptor-alpha and -beta, progesterone receptor, and androgen receptor mRNA in normal and malignant ovarian epithelial cells. Proc Natl Acad Sci USA 96:5722–5727CrossRefPubMed Lau KM, Mok SC, Ho SM (1999) Expression of human estrogen receptor-alpha and -beta, progesterone receptor, and androgen receptor mRNA in normal and malignant ovarian epithelial cells. Proc Natl Acad Sci USA 96:5722–5727CrossRefPubMed
21.
go back to reference Thomas P (2008) Characteristics of membrane progestin receptor alpha (mPRalpha) and progesterone membrane receptor component 1 (PGMRC1) and their roles in mediating rapid progestin actions. Front Neuroendocrinol 29:292–312CrossRefPubMed Thomas P (2008) Characteristics of membrane progestin receptor alpha (mPRalpha) and progesterone membrane receptor component 1 (PGMRC1) and their roles in mediating rapid progestin actions. Front Neuroendocrinol 29:292–312CrossRefPubMed
22.
go back to reference Zhu Y, Bond J, Thomas P (2003) Identification, classification, and partial characterization of genes in humans and other vertebrates homologous to a fish membrane progestin receptor. Proc Natl Acad Sci USA 100:2237–2242CrossRefPubMed Zhu Y, Bond J, Thomas P (2003) Identification, classification, and partial characterization of genes in humans and other vertebrates homologous to a fish membrane progestin receptor. Proc Natl Acad Sci USA 100:2237–2242CrossRefPubMed
23.
go back to reference Zhu Y, Rice CD, Pang Y, Pace M, Thomas P (2003) Cloning, expression, and characterization of a membrane progestin receptor and evidence it is an intermediary in meiotic maturation of fish oocytes. Proc Natl Acad Sci USA 100:2231–2236CrossRefPubMed Zhu Y, Rice CD, Pang Y, Pace M, Thomas P (2003) Cloning, expression, and characterization of a membrane progestin receptor and evidence it is an intermediary in meiotic maturation of fish oocytes. Proc Natl Acad Sci USA 100:2231–2236CrossRefPubMed
24.
go back to reference Thomas P, Pang Y, Dong J, Groenen P, Kelder J, de Vlieg J et al (2007) Steroid and G protein binding characteristics of the seatrout and human progestin membrane receptor alpha subtypes and their evolutionary origins. Endocrinology 148:705–718CrossRefPubMed Thomas P, Pang Y, Dong J, Groenen P, Kelder J, de Vlieg J et al (2007) Steroid and G protein binding characteristics of the seatrout and human progestin membrane receptor alpha subtypes and their evolutionary origins. Endocrinology 148:705–718CrossRefPubMed
25.
go back to reference Karteris E, Zervou S, Pang Y, Dong J, Hillhouse EW, Randeva HS et al (2006) Progesterone signaling in human myometrium through two novel membrane G protein-coupled receptors: potential role in functional progesterone withdrawal at term. Mol Endocrinol 20:1519–1534CrossRefPubMed Karteris E, Zervou S, Pang Y, Dong J, Hillhouse EW, Randeva HS et al (2006) Progesterone signaling in human myometrium through two novel membrane G protein-coupled receptors: potential role in functional progesterone withdrawal at term. Mol Endocrinol 20:1519–1534CrossRefPubMed
26.
go back to reference Tubbs C, Thomas P (2009) Progestin signaling through an olfactory G protein and membrane progestin receptor-alpha in Atlantic croaker sperm: potential role in induction of sperm hypermotility. Endocrinology 150:473–484CrossRefPubMed Tubbs C, Thomas P (2009) Progestin signaling through an olfactory G protein and membrane progestin receptor-alpha in Atlantic croaker sperm: potential role in induction of sperm hypermotility. Endocrinology 150:473–484CrossRefPubMed
27.
go back to reference Cai Z, Stocco C (2005) Expression and regulation of progestin membrane receptors in the rat corpus luteum. Endocrinology 146:5522–5532CrossRefPubMed Cai Z, Stocco C (2005) Expression and regulation of progestin membrane receptors in the rat corpus luteum. Endocrinology 146:5522–5532CrossRefPubMed
28.
go back to reference Ashley RL, Arreguin-Arevalo JA, Nett TM (2009) Binding characteristics of the ovine membrane progesterone receptor alpha and expression of the receptor during the estrous cycle. Reprod Biol Endocrinol 7:42CrossRefPubMed Ashley RL, Arreguin-Arevalo JA, Nett TM (2009) Binding characteristics of the ovine membrane progesterone receptor alpha and expression of the receptor during the estrous cycle. Reprod Biol Endocrinol 7:42CrossRefPubMed
29.
go back to reference Ashley RL, Clay CM, Farmerie TA, Niswender GD, Nett TM (2006) Cloning and characterization of an ovine intracellular seven transmembrane receptor for progesterone that mediates calcium mobilization. Endocrinology 147:4151–4159CrossRefPubMed Ashley RL, Clay CM, Farmerie TA, Niswender GD, Nett TM (2006) Cloning and characterization of an ovine intracellular seven transmembrane receptor for progesterone that mediates calcium mobilization. Endocrinology 147:4151–4159CrossRefPubMed
30.
go back to reference Romero-Sanchez M, Peiper SC, Evans B, Wang Z, Catasus L, Ribe A et al (2008) Expression profile of heptahelical putative membrane progesterone receptors in epithelial ovarian tumors. Hum Pathol 39:1026–1033CrossRefPubMed Romero-Sanchez M, Peiper SC, Evans B, Wang Z, Catasus L, Ribe A et al (2008) Expression profile of heptahelical putative membrane progesterone receptors in epithelial ovarian tumors. Hum Pathol 39:1026–1033CrossRefPubMed
31.
go back to reference Boulware MI, Weick JP, Becklund BR, Kuo SP, Groth RD, Mermelstein PG (2005) Estradiol activates group I and II metabotropic glutamate receptor signaling, leading to opposing influences on cAMP response element-binding protein. J Neurosci 25:5066–5078CrossRefPubMed Boulware MI, Weick JP, Becklund BR, Kuo SP, Groth RD, Mermelstein PG (2005) Estradiol activates group I and II metabotropic glutamate receptor signaling, leading to opposing influences on cAMP response element-binding protein. J Neurosci 25:5066–5078CrossRefPubMed
32.
go back to reference Pierson-Mullany LK, Lange CA (2004) Phosphorylation of progesterone receptor serine 400 mediates ligand-independent transcriptional activity in response to activation of cyclin-dependent protein kinase 2. Mol Cell Biol 24:10542–10557CrossRefPubMed Pierson-Mullany LK, Lange CA (2004) Phosphorylation of progesterone receptor serine 400 mediates ligand-independent transcriptional activity in response to activation of cyclin-dependent protein kinase 2. Mol Cell Biol 24:10542–10557CrossRefPubMed
33.
go back to reference Dressing GE, Thomas P (2007) Identification of membrane progestin receptors in human breast cancer cell lines and biopsies and their potential involvement in breast cancer. Steroids 72:111–116CrossRefPubMed Dressing GE, Thomas P (2007) Identification of membrane progestin receptors in human breast cancer cell lines and biopsies and their potential involvement in breast cancer. Steroids 72:111–116CrossRefPubMed
34.
go back to reference Milligan G (2009) G protein-coupled receptor hetero-dimerization: contribution to pharmacology and function. Br J Pharmacol 158:5–14CrossRefPubMed Milligan G (2009) G protein-coupled receptor hetero-dimerization: contribution to pharmacology and function. Br J Pharmacol 158:5–14CrossRefPubMed
35.
go back to reference Shaw TJ, Keszthelyi EJ, Tonary AM, Cada M, Vanderhyden BC (2002) Cyclic AMP in ovarian cancer cells both inhibits proliferation and increases c-KIT expression. Exp Cell Res 273:95–106CrossRefPubMed Shaw TJ, Keszthelyi EJ, Tonary AM, Cada M, Vanderhyden BC (2002) Cyclic AMP in ovarian cancer cells both inhibits proliferation and increases c-KIT expression. Exp Cell Res 273:95–106CrossRefPubMed
36.
go back to reference Mayr B, Montminy M (2001) Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat Rev Mol Cell Biol 2:599–609CrossRefPubMed Mayr B, Montminy M (2001) Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat Rev Mol Cell Biol 2:599–609CrossRefPubMed
37.
go back to reference Syed V, Mukherjee K, Lyons-Weiler J, Lau KM, Mashima T, Tsuruo T et al (2005) Identification of ATF-3, caveolin-1, DLC-1, and NM23-H2 as putative antitumorigenic, progesterone-regulated genes for ovarian cancer cells by gene profiling. Oncogene 24:1774–1787CrossRefPubMed Syed V, Mukherjee K, Lyons-Weiler J, Lau KM, Mashima T, Tsuruo T et al (2005) Identification of ATF-3, caveolin-1, DLC-1, and NM23-H2 as putative antitumorigenic, progesterone-regulated genes for ovarian cancer cells by gene profiling. Oncogene 24:1774–1787CrossRefPubMed
38.
go back to reference Matsukawa J, Matsuzawa A, Takeda K, Ichijo H (2004) The ASK1-MAP kinase cascades in mammalian stress response. J Biochem 136:261–265CrossRefPubMed Matsukawa J, Matsuzawa A, Takeda K, Ichijo H (2004) The ASK1-MAP kinase cascades in mammalian stress response. J Biochem 136:261–265CrossRefPubMed
39.
go back to reference Miller WE, Lefkowitz RJ (2001) Expanding roles for beta-arrestins as scaffolds and adapters in GPCR signaling and trafficking. Curr Opin Cell Biol 13:139–145CrossRefPubMed Miller WE, Lefkowitz RJ (2001) Expanding roles for beta-arrestins as scaffolds and adapters in GPCR signaling and trafficking. Curr Opin Cell Biol 13:139–145CrossRefPubMed
40.
go back to reference Arafat WO, Gomez-Navarro J, Xiang J, Barnes MN, Mahasreshti P, Alvarez RD et al (2000) An adenovirus encoding proapoptotic Bax induces apoptosis and enhances the radiation effect in human ovarian cancer. Mol Ther 1:545–554CrossRefPubMed Arafat WO, Gomez-Navarro J, Xiang J, Barnes MN, Mahasreshti P, Alvarez RD et al (2000) An adenovirus encoding proapoptotic Bax induces apoptosis and enhances the radiation effect in human ovarian cancer. Mol Ther 1:545–554CrossRefPubMed
41.
go back to reference Marone M, Scambia G, Mozzetti S, Ferrandina G, Iacovella S, De Pasqua A et al (1998) bcl-2, bax, bcl-XL, and bcl-XS expression in normal and neoplastic ovarian tissues. Clin Cancer Res 4:517–524PubMed Marone M, Scambia G, Mozzetti S, Ferrandina G, Iacovella S, De Pasqua A et al (1998) bcl-2, bax, bcl-XL, and bcl-XS expression in normal and neoplastic ovarian tissues. Clin Cancer Res 4:517–524PubMed
42.
go back to reference Wu GS (2004) The functional interactions between the p53 and MAPK signaling pathways. Cancer Biol Ther 3:156–161PubMed Wu GS (2004) The functional interactions between the p53 and MAPK signaling pathways. Cancer Biol Ther 3:156–161PubMed
43.
go back to reference Perego P, Giarola M, Righetti SC, Supino R, Caserini C, Delia D et al (1996) Association between cisplatin resistance and mutation of p53 gene and reduced bax expression in ovarian carcinoma cell systems. Cancer Res 56:556–562PubMed Perego P, Giarola M, Righetti SC, Supino R, Caserini C, Delia D et al (1996) Association between cisplatin resistance and mutation of p53 gene and reduced bax expression in ovarian carcinoma cell systems. Cancer Res 56:556–562PubMed
44.
go back to reference Fister S, Gunthert AR, Aicher B, Paulini KW, Emons G, Grundker C (2009) GnRH-II antagonists induce apoptosis in human endometrial, ovarian, and breast cancer cells via activation of stress-induced MAPKs p38 and JNK and proapoptotic protein Bax. Cancer Res 69:6473–6481CrossRefPubMed Fister S, Gunthert AR, Aicher B, Paulini KW, Emons G, Grundker C (2009) GnRH-II antagonists induce apoptosis in human endometrial, ovarian, and breast cancer cells via activation of stress-induced MAPKs p38 and JNK and proapoptotic protein Bax. Cancer Res 69:6473–6481CrossRefPubMed
45.
go back to reference Kim BJ, Ryu SW, Song BJ (2006) JNK- and p38 kinase-mediated phosphorylation of Bax leads to its activation and mitochondrial translocation and to apoptosis of human hepatoma HepG2 cells. J Biol Chem 281:21256–21265CrossRefPubMed Kim BJ, Ryu SW, Song BJ (2006) JNK- and p38 kinase-mediated phosphorylation of Bax leads to its activation and mitochondrial translocation and to apoptosis of human hepatoma HepG2 cells. J Biol Chem 281:21256–21265CrossRefPubMed
46.
go back to reference Mansouri A, Ridgway LD, Korapati AL, Zhang Q, Tian L, Wang Y et al (2003) Sustained activation of JNK/p38 MAPK pathways in response to cisplatin leads to Fas ligand induction and cell death in ovarian carcinoma cells. J Biol Chem 278:19245–19256CrossRefPubMed Mansouri A, Ridgway LD, Korapati AL, Zhang Q, Tian L, Wang Y et al (2003) Sustained activation of JNK/p38 MAPK pathways in response to cisplatin leads to Fas ligand induction and cell death in ovarian carcinoma cells. J Biol Chem 278:19245–19256CrossRefPubMed
47.
go back to reference Lee LF, Li G, Templeton DJ, Ting JP (1998) Paclitaxel (Taxol)-induced gene expression and cell death are both mediated by the activation of c-Jun NH2-terminal kinase (JNK/SAPK). J Biol Chem 273:28253–28260CrossRefPubMed Lee LF, Li G, Templeton DJ, Ting JP (1998) Paclitaxel (Taxol)-induced gene expression and cell death are both mediated by the activation of c-Jun NH2-terminal kinase (JNK/SAPK). J Biol Chem 273:28253–28260CrossRefPubMed
48.
go back to reference Givant-Horwitz V, Davidson B, Lazarovici P, Schaefer E, Nesland JM, Trope CG et al (2003) Mitogen-activated protein kinases (MAPK) as predictors of clinical outcome in serous ovarian carcinoma in effusions. Gynecol Oncol 91:160–172CrossRefPubMed Givant-Horwitz V, Davidson B, Lazarovici P, Schaefer E, Nesland JM, Trope CG et al (2003) Mitogen-activated protein kinases (MAPK) as predictors of clinical outcome in serous ovarian carcinoma in effusions. Gynecol Oncol 91:160–172CrossRefPubMed
49.
go back to reference Bu SZ, Yin DL, Ren XH, Jiang LZ, Wu ZJ, Gao QR et al (1997) Progesterone induces apoptosis and up-regulation of p53 expression in human ovarian carcinoma cell lines. Cancer 79:1944–1950CrossRefPubMed Bu SZ, Yin DL, Ren XH, Jiang LZ, Wu ZJ, Gao QR et al (1997) Progesterone induces apoptosis and up-regulation of p53 expression in human ovarian carcinoma cell lines. Cancer 79:1944–1950CrossRefPubMed
50.
go back to reference Syed V, Ho SM (2003) Progesterone-induced apoptosis in immortalized normal and malignant human ovarian surface epithelial cells involves enhanced expression of FasL. Oncogene 22:6883–6890CrossRefPubMed Syed V, Ho SM (2003) Progesterone-induced apoptosis in immortalized normal and malignant human ovarian surface epithelial cells involves enhanced expression of FasL. Oncogene 22:6883–6890CrossRefPubMed
51.
go back to reference Syed V, Mukherjee K, Godoy-Tundidor S, Ho SM (2007) Progesterone induces apoptosis in TRAIL-resistant ovarian cancer cells by circumventing c-FLIPL overexpression. J Cell Biochem 102:442–452CrossRefPubMed Syed V, Mukherjee K, Godoy-Tundidor S, Ho SM (2007) Progesterone induces apoptosis in TRAIL-resistant ovarian cancer cells by circumventing c-FLIPL overexpression. J Cell Biochem 102:442–452CrossRefPubMed
52.
go back to reference Chao KC, Wang PH, Yen MS, Chang CC, Chi CW (2005) Role of estrogen and progesterone in the survival of ovarian tumors–a study of the human ovarian adenocarcinoma cell line OC-117-VGH. J Chin Med Assoc 68:360–367CrossRefPubMed Chao KC, Wang PH, Yen MS, Chang CC, Chi CW (2005) Role of estrogen and progesterone in the survival of ovarian tumors–a study of the human ovarian adenocarcinoma cell line OC-117-VGH. J Chin Med Assoc 68:360–367CrossRefPubMed
53.
go back to reference Belinson JL, McClure M, Badger G (1987) Randomized trial of megestrol acetate vs. megestrol acetate/tamoxifen for the management of progressive or recurrent epithelial ovarian carcinoma. Gynecol Oncol 28:151–155CrossRefPubMed Belinson JL, McClure M, Badger G (1987) Randomized trial of megestrol acetate vs. megestrol acetate/tamoxifen for the management of progressive or recurrent epithelial ovarian carcinoma. Gynecol Oncol 28:151–155CrossRefPubMed
54.
go back to reference Chen X, Feng Y (2003) Effect of progesterone combined with chemotherapy on epithelial ovarian cancer. Chin Med J (Engl) 116:388–391 Chen X, Feng Y (2003) Effect of progesterone combined with chemotherapy on epithelial ovarian cancer. Chin Med J (Engl) 116:388–391
Metadata
Title
Expression of Membrane Progesterone Receptors (mPR/PAQR) in Ovarian Cancer Cells: Implications for Progesterone-Induced Signaling Events
Authors
Nathan J. Charles
Peter Thomas
Carol A. Lange
Publication date
01-08-2010
Publisher
Springer-Verlag
Published in
Discover Oncology / Issue 4/2010
Print ISSN: 1868-8497
Electronic ISSN: 2730-6011
DOI
https://doi.org/10.1007/s12672-010-0023-9

Other articles of this Issue 4/2010

Discover Oncology 4/2010 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine