Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Semin Reprod Med ; 28(1): 44-50, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20108182

ABSTRACT

Estradiol (E2) stimulates the growth and inflammation in the ectopic endometriotic tissue that commonly resides on the pelvic organs. Several clinical and laboratory-based observations are indicative of resistance to progesterone action in endometriosis. The molecular basis of progesterone resistance in endometriosis may be related to an overall reduction in the levels of progesterone receptor (PR). In normal endometrium, progesterone acts via PR on stromal cells to induce secretion of paracrine factor(s) that in turn stimulate neighboring epithelial cells to express the enzyme 17beta-hydroxysteroid dehydrogenase type 2 (HSD17B2). HSD17B2 is an extremely efficient enzyme and rapidly metabolizes the biologically potent estrogen E2 to weakly estrogenic estrone. In endometriotic tissue, progesterone is incapable of inducing epithelial HSD17B2 expression due to a defect in stromal cells. The inability of endometriotic stromal cells to produce progesterone-induced paracrine factors that stimulate HSD17B2 may be due to the very low levels of PR observed in vivo in endometriotic tissue. The end result is deficient metabolism of E2 in endometriosis giving rise to high local concentrations of this mitogen. The molecular details of this physiological paracrine interaction between the stroma and epithelium in normal endometrium and its lack thereof in endometriosis are discussed.


Subject(s)
17-Hydroxysteroid Dehydrogenases/deficiency , Endometriosis/enzymology , Progesterone/metabolism , Animals , Cell Differentiation/genetics , Endometriosis/metabolism , Female , Gene Expression Regulation , Humans , Mice , Paracrine Communication , Receptors, Progesterone/deficiency , Receptors, Progesterone/metabolism , Stromal Cells/metabolism
2.
Semin Reprod Med ; 28(1): 36-43, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20104427

ABSTRACT

Loss of progesterone signaling in the endometrium may be a causal factor in the development of endometriosis, and progesterone resistance is commonly observed in women with this disease. In endometriotic stromal cells, the levels of progesterone receptor (PR), particularly the PR-B isoform, are significantly decreased, leading to a loss of paracrine signaling. PR deficiency likely underlies the development of progesterone resistance in women with endometriosis who no longer respond to progestin therapy. Here we review the complex epigenetic and transcriptional mechanisms leading to PR deficiency. The initial event may involve deficient methylation of the estrogen receptor (ER)beta promoter resulting in pathologic overexpression of ERbeta in endometriotic stromal cells. We speculate that alterations in the relative levels of ERbeta and ERalpha in endometrial tissue dictate E2-regulated PR expression, such that a decreased ERalpha-tauomicron-ERbeta ratio may result in suppression of PR. In this review, we propose a molecular model that may be responsible for changes in ERbeta and ERalpha leading to PR loss and progesterone resistance in endometriosis.


Subject(s)
Endometriosis/metabolism , Epigenesis, Genetic , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Gene Expression Regulation, Neoplastic , Progesterone/metabolism , CpG Islands , DNA Methylation , Endometriosis/genetics , Endometrium/metabolism , Female , Humans , Receptors, Progesterone/genetics , Stromal Cells/metabolism , Stromal Cells/pathology
3.
J Clin Endocrinol Metab ; 94(2): 623-31, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19001523

ABSTRACT

CONTEXT: Products of at least five specific steroidogenic genes, including steroidogenic acute regulatory protein (StAR), which facilitates the entry of cytosolic cholesterol into the mitochondrion, side chain cleavage P450 enzyme, 3beta-hydroxysteroid-dehydrogenase-2, 17-hydroxylase/17-20-lyase, and aromatase, which catalyzes the final step, are necessary for the conversion of cholesterol to estrogen. Expression and biological activity of StAR and aromatase were previously demonstrated in endometriosis but not in normal endometrium. Prostaglandin E2 (PGE2) induces aromatase expression via the transcriptional factor steroidogenic factor-1 (SF1) in endometriosis, which is opposed by chicken-ovalbumin upstream-transcription factor (COUP-TF) and Wilms' tumor-1 (WT1) in endometrium. OBJECTIVE: The aim of the study was to demonstrate a complete steroidogenic pathway leading to estrogen biosynthesis in endometriotic cells and the transcriptional mechanisms that regulate basal and PGE2-stimulated estrogen production in endometriotic cells and endometrium. RESULTS: Compared with normal endometrial tissues, mRNA levels of StAR, side chain cleavage P450, 3beta-hydroxysteroid-dehydrogenase-2, 17-hydroxylase/17-20-lyase, aromatase, and SF1 were significantly higher in endometriotic tissues. PGE2 induced the expression of all steroidogenic genes; production of progesterone, estrone, and estradiol; and StAR promoter activity in endometriotic cells. Overexpression of SF1 induced, whereas COUP-TFII or WT1 suppressed, StAR promoter activity. PGE2 induced coordinate binding of SF1 to StAR and aromatase promoters but decreased COUP-TFII binding in endometriotic cells. COUP-TFII or WT1 binding to both promoters was significantly higher in endometrial compared with endometriotic cells. CONCLUSION: Endometriotic cells contain the full complement of steroidogenic genes for de novo synthesis of estradiol from cholesterol, which is stimulated by PGE2 via enhanced binding of SF1 to promoters of StAR and aromatase genes in a synchronous fashion.


Subject(s)
Dinoprostone/pharmacology , Endometriosis/genetics , Estradiol/biosynthesis , Gene Expression Regulation, Enzymologic , Ovarian Diseases/genetics , Steroidogenic Factor 1/physiology , Adult , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Endometriosis/enzymology , Endometriosis/metabolism , Endometrium/drug effects , Endometrium/metabolism , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Middle Aged , Ovarian Diseases/enzymology , Ovarian Diseases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Progesterone/biosynthesis , Promoter Regions, Genetic/drug effects , Transcription, Genetic/drug effects , Young Adult
4.
Mol Endocrinol ; 22(4): 904-14, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18165439

ABSTRACT

Local estrogen biosynthesis is a major factor in the pathogenesis of endometriosis. Aberrant expression of steroidogenic acute regulatory protein (StAR) and aromatase in endometriotic tissue leads to an up-regulation of estrogen production. The transcription factor steroidogenic factor-1 (SF-1) activates the promoters of both StAR and aromatase in endometriotic tissue. We investigated differences in SF-1 expression in endometriotic tissue and normally located endometrium to elucidate the mechanism underlying increased StAR and aromatase activities in endometriosis. Serial deletion and site-directed mutants of the SF-1 promoter showed that an E-box sequence was critical for its activity in endometriotic stromal cells. EMSAs showed that the upstream stimulatory factor (USF) 1 and 2 in nuclear extracts from endometrial and endometriotic stromal cells bound to the E-box. Chromatin-immunoprecipitation-PCR assay, however, demonstrated in intact cells that binding activity of USF2 to the SF-1 promoter was strikingly higher than that of USF1 in endometriotic stromal cells and that USF1 or USF2 binding activity was hardly detectable in endometrial stromal cells. Moreover, knockdown of USF2 but not USF1 resulted in robust and consistent down-regulation of SF-1 and its target genes StAR and aromatase in endometriotic stromal cells. USF2 but not USF1 mRNA and protein levels were significantly higher in endometriotic vs. endometrial stromal cells. In vivo, USF2 mRNA and immunoreactive USF2 levels in endometriotic tissues were strikingly higher than those in endometrium. Taken together, the elevated levels of USF2 in endometriosis account for, in part, the aberrant expression of SF-1 and its target gene StAR and aromatase.


Subject(s)
Endometriosis/genetics , Gene Expression , Steroidogenic Factor 1/genetics , Upstream Stimulatory Factors/physiology , Aromatase/genetics , Aromatase/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/metabolism , Endometrium/pathology , Female , Humans , Immunoblotting , Immunohistochemistry , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroidogenic Factor 1/metabolism , Stromal Cells/metabolism , Transcription, Genetic/genetics , Transfection , Upstream Stimulatory Factors/genetics , Upstream Stimulatory Factors/metabolism
5.
J Steroid Biochem Mol Biol ; 106(1-5): 81-96, 2007.
Article in English | MEDLINE | ID: mdl-17590327

ABSTRACT

Pathogenesis and growth of three common women's cancers (breast, endometrium and ovary) are linked to estrogen. A single gene encodes the key enzyme for estrogen biosynthesis named aromatase, inhibition of which effectively eliminates estrogen production in the entire body. Aromatase inhibitors successfully treat breast cancer, whereas their roles in endometrial and ovarian cancers are less clear. Ovary, testis, adipose tissue, skin, hypothalamus and placenta express aromatase normally, whereas breast, endometrial and ovarian cancers overexpress aromatase and produce local estrogen exerting paracrine and intracrine effects. Tissue-specific promoters distributed over a 93-kb regulatory region upstream of a common coding region alternatively control aromatase expression. A distinct set of transcription factors regulates each promoter in a signaling pathway- and tissue-specific manner. In cancers of breast, endometrium and ovary, aromatase expression is primarly regulated by increased activity of the proximally located promoter I.3/II region. Promoters I.3 and II lie 215 bp from each other and are coordinately stimulated by PGE(2) via a cAMP-PKA-dependent pathway. In breast adipose fibroblasts exposed to PGE(2) secreted by malignant epithelial cells, PKC is also activated, and this potentiates cAMP-PKA-dependent induction of aromatase. Thus, inflammatory substances such as PGE(2) may play important roles in inducing local production of estrogen that promotes tumor growth.


Subject(s)
Aromatase/metabolism , Breast Neoplasms/enzymology , Endometrial Neoplasms/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/enzymology , Animals , Aromatase/genetics , Breast Neoplasms/genetics , Endometrial Neoplasms/genetics , Female , Humans , Ovarian Neoplasms/genetics
6.
J Obstet Gynaecol Res ; 32(6): 613-4, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17100826

ABSTRACT

A 30-year-old woman with a history of endometriosis and chronic pelvic pain had right-sided pain and sonographic evaluation demonstrated a right ovarian cyst 5 cm in diameter. Laparotomy revealed a right ovarian cystic mass and the cystic mass was found on the sigmoid colon. After excision, histopathologic study revealed endometrioma for the ovarian cyst and a supernumerary ovary for the cystic mass on the sigmoid colon.


Subject(s)
Choristoma , Ovarian Cysts/complications , Ovary , Pelvic Pain/etiology , Sigmoid Diseases/pathology , Adult , Female , Histocytochemistry , Humans
7.
Mol Cell Endocrinol ; 248(1-2): 94-103, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16406281

ABSTRACT

Endometriosis is the most common cause of pelvic pain and affects an estimated 5 million women in the US. The biologically active estrogen estradiol (E2) is the best-defined mitogen for the growth and inflammation processes in the ectopic endometriotic tissue that commonly resides on the pelvic organs. Progesterone and progestins may relieve pain by limiting growth and inflammation in endometriosis but a portion of patients with endometriosis and pelvic pain do not respond to treatment with progestins. Moreover, progesterone-induced molecular changes in the eutopic (intrauterine) endometrial tissue of women with endometriosis are either blunted or undetectable. These in vivo observations are indicative of resistance to progesterone action in endometriosis. The molecular basis of progesterone resistance in endometriosis may be related to an overall reduction in the levels of progesterone receptors (PRs) and the lack of the PR isoform named progesterone receptor B (PR-B). In normal endometrium, progesterone acts on stromal cells to induce secretion of paracrine factor(s). These unknown factor(s) act on neighboring epithelial cells to induce the expression of the enzyme 17beta-hydroxysteroid dehydrogenase type 2 (17beta-HSD-2), which metabolizes the biologically active estrogen E2 to estrone (E1). In endometriotic tissue, progesterone does not induce epithelial 17beta-HSD-2 expression due to a defect in stromal cells. The inability of endometriotic stromal cells to produce progesterone-induced paracrine factors that stimulate 17beta-HSD-2 may be due to the lack of PR-B and very low levels of progesterone receptor A (PR-A) observed in vivo in endometriotic tissue. The end result is deficient metabolism of E2 in endometriosis giving rise to high local concentrations of this local mitogen. The cellular and molecular mechanisms underlying progesterone resistance and failure to metabolize E2 in endometriosis are reviewed.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , Endometriosis/metabolism , Genital Diseases, Female/metabolism , Progesterone/physiology , Receptors, Progesterone/deficiency , Drug Resistance/genetics , Endometriosis/drug therapy , Estradiol/metabolism , Estradiol Dehydrogenases , Female , Genital Diseases, Female/drug therapy , Humans , Progesterone/therapeutic use
8.
Pharmacol Rev ; 57(3): 359-83, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16109840

ABSTRACT

A single gene encodes the key enzyme for estrogen biosynthesis termed aromatase, inhibition of which effectively eliminates estrogen production. Aromatase inhibitors successfully treat breast cancer and endometriosis, whereas their roles in endometrial cancer, uterine fibroids, and aromatase excess syndrome are less clear. Ovary, testis, adipose tissue, skin, hypothalamus, and placenta express aromatase normally, whereas breast and endometrial cancers, endometriosis, and uterine fibroids overexpress aromatase and produce local estrogen that exerts paracrine and intracrine effects. Tissue-specific promoters distributed over a 93-kilobase regulatory region upstream of a common coding region alternatively control aromatase expression. A distinct set of transcription factors regulates each promoter in a signaling pathway- and tissue-specific manner. Three mechanisms are responsible for aromatase overexpression in a pathologic tissue versus its normal counterpart. First, cellular composition is altered to increase aromatase-expressing cell types that use distinct promoters (breast cancer). Second, molecular alterations in stromal cells favor binding of transcriptional enhancers versus inhibitors to a normally quiescent aromatase promoter and initiate transcription (breast/endometrial cancer, endometriosis, and uterine fibroids). Third, heterozygous mutations, which cause the aromatase coding region to lie adjacent to constitutively active cryptic promoters that normally transcribe other genes, result in excessive estrogen formation owing to the overexpression of aromatase in many tissues.


Subject(s)
Aromatase/biosynthesis , Breast Neoplasms/enzymology , Estrogens/metabolism , Gene Expression Regulation, Enzymologic , Ovarian Diseases/enzymology , Animals , Aromatase/genetics , Aromatase Inhibitors/pharmacology , Breast Neoplasms/metabolism , Female , Humans , Ovarian Diseases/metabolism , Promoter Regions, Genetic
9.
J Steroid Biochem Mol Biol ; 95(1-5): 57-62, 2005 May.
Article in English | MEDLINE | ID: mdl-16024248

ABSTRACT

Endometrial tissue from uterine disease-free women does not exhibit aromatase activity. In contrast, aromatase enzyme activity and mRNA levels are readily detectable in endometriosis. PGE2 stimulates both aromatase expression and activity in endometriotic stromal cells via promoter II region of the aromatase gene. This results in local production of estradiol, which induces PGE2 formation and establishes a positive feedback cycle. This mechanism seems to contribute to continuous production of estradiol and PGE2. Aromatase mRNA levels and enzyme activity are also present in uterine leiomyomata that are estrogen-dependent benign tumors of the myometrium. Successful treatment of endometriosis and uterine leiomyomata using aromatase inhibitors by recent pilot trials underscores the clinical significance of these molecular studies.


Subject(s)
Aromatase/metabolism , Endometriosis/enzymology , Leiomyoma/enzymology , Uterine Neoplasms/enzymology , Aromatase/drug effects , Aromatase/genetics , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Endometriosis/drug therapy , Estrogens/metabolism , Female , Humans , Uterine Neoplasms/drug therapy , Uterus/enzymology , Uterus/pathology
10.
Semin Reprod Med ; 22(1): 45-50, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15083380

ABSTRACT

Aromatase p450 (p450arom) is the key enzyme for biosynthesis of estrogen, which is an essential hormone for the establishment and growth of endometriosis. There is no detectable aromatase enzyme activity in normal endometrium; therefore, estrogen is not locally produced in endometrium. Endometriosis tissue, however, contains very high levels of aromatase enzyme, which leads to production of significant quantities of estrogen. Moreover, one of the best-known mediators of inflammation and pain, prostaglandin E (2), strikingly induces aromatase enzyme activity and formation of local estrogen in this tissue. Additionally, estrogen itself stimulates cyclo-oxygenase-2 and therefore increases the formation of prostaglandin E (2) in endometriosis. We were able to target this positive feedback cycle in endometriosis using aromatase inhibitors. In fact, pilot trials showed that aromatase inhibitors could decrease pelvic pain associated with endometriosis.


Subject(s)
Aromatase/metabolism , Endometriosis/enzymology , Endometriosis/drug therapy , Enzyme Inhibitors/therapeutic use , Estrogens/biosynthesis , Female , Gene Expression Regulation , Humans , Stromal Cells/enzymology
11.
Hum Reprod ; 17(7): 1729-37, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12093831

ABSTRACT

BACKGROUND: Oral contraceptive pills (OC) are usually the first choice of treatment for polycystic ovarian syndrome (PCOS), when fertility is not desired. However, they do not improve, or may even further induce impairment of insulin sensitivity, which is already impaired in women with PCOS. In this prospective, randomized study, we analysed the additional benefits of adding metformin to the OC treatment in non-obese women with PCOS. METHODS: After a baseline work-up including body mass index (BMI), waist:hip ratio (WHR), Ferriman-Gallwey score, ovarian volume, serum gonadotrophin, androgen and sex hormone-binding globulin (SHBG) levels, and fasting lipid, glucose and insulin levels, 40 non-obese women with PCOS were assigned either to the OC or to the OC + metformin treatment by computer-assisted randomization. At the end of the 4 month follow-up period, subjects were re-evaluated. RESULTS: The two groups were similar at baseline. After treatment, women in the OC + metformin group had significant decreases in BMI and WHR, and a significant increase in insulin sensitivity, in contrast to those in the OC group, who had insignificant changes in these parameters. Adding metformin also caused significant improvements in serum androstenedione and SHBG levels compared with the OC treatment alone. CONCLUSIONS: Adding metformin to the OC treatment may improve the insulin sensitivity, and may further suppress the hyperandrogenaemia in non-obese women with PCOS.


Subject(s)
Cyproterone Acetate/therapeutic use , Ethinyl Estradiol/therapeutic use , Metformin/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Adolescent , Adult , Androstenedione/blood , Body Constitution , Body Mass Index , Drug Therapy, Combination , Female , Humans , Insulin Resistance , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/pathology , Prospective Studies , Sex Hormone-Binding Globulin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...