Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cancer Sci ; 99(2): 267-71, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18271925

ABSTRACT

The retinoids (vitamin A and its biologically active derivatives) are essential for the health and survival of the individual. Several studies have reported a strong rationale for the use of retinoids in cancer treatment and chemoprevention. It has been discovered that expression of retinoic acid receptor (RAR) beta is frequently silenced in epithelial carcinogenesis, which has led to the hypothesis that RAR beta could act as a tumor suppressor. However, the status of RAR beta in human endometrial carcinoma has not been examined. In the present study, we initially studied the effects of retinoic acid on cell proliferation and the expression of RAR alpha, RAR beta, and RAR gamma using AM580 (a RAR-specific agonist) in the Ishikawa endometrial cancer cell line. We also examined the expression of RAR in human eutopic endometrium (30 cases), endometrial hyperplasia (28 cases), and endometrial carcinoma (103 cases) using immunohistochemistry. Finally, we correlated these findings with the clinicopathological parameters. In vitro, cell growth was inhibited and RAR beta and RAR gamma mRNA was significantly induced by AM580, compared with vehicle controls, whereas RAR alpha mRNA was significantly attenuated by AM580, compared with vehicle. RAR beta was detected predominantly in endometrial hyperplasia, compared with endometrial carcinoma. No statistically significant correlation was obtained between the expression of any other RAR subtypes and clinicopathological parameters in human endometrial carcinoma. The results of our study demonstrate that AM580 inhibits cell growth and induces RAR beta mRNA expression in the Ishikawa cell line, and the expression level of RAR beta in endometrial carcinoma is significantly lower than that in endometrial hyperplasia. AM580 might therefore be considered as a potential treatment for endometrial carcinoma.


Subject(s)
Carcinoma/metabolism , Endometrial Neoplasms/metabolism , Receptors, Retinoic Acid/metabolism , Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Carcinoma/genetics , Carcinoma/pathology , Cell Proliferation , Endometrial Hyperplasia/metabolism , Endometrial Hyperplasia/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrium/metabolism , Female , Gene Expression , Humans , RNA, Messenger/metabolism , Receptors, Retinoic Acid/analysis , Receptors, Retinoic Acid/genetics , Tetrahydronaphthalenes/pharmacology , Tretinoin/pharmacology , Tumor Cells, Cultured
2.
J Minim Invasive Gynecol ; 15(1): 74-7, 2008.
Article in English | MEDLINE | ID: mdl-18262148

ABSTRACT

STUDY OBJECTIVE: To analyze variables for successful 1-step hysteroscopic myomectomies of sessile submucous myomas. DESIGN: Retrospective case-control study. (Canadian Task Force classification II-2). SETTING: Single operator's practice in a university hospital and its related hospitals. PATIENTS: Twenty-eight patients with sessile submucous myomas and menorrhagia, infertility, or both. INTERVENTIONS: Our strategy for hysteroscopic myomectomy is as follows. First, we scraped and/or vaporized intrauterine dome of myoma until top of myoma was even with level of wall of cavity. Next, the remnant intramural node was squeezed by uterine contractions induced by prostaglandin F2alpha injection. Finally, the newly raised myoma dome was sectioned or vaporized electrosurgically only within the space of the intrauterine cavity and/or was separated mechanically from healthy myometrium without electrosurgery. MEASUREMENTS AND MAIN RESULTS: Submucous myomas in 16 (57.1%) patients were completely removed after 1 surgery. By logistic regression analysis, thickness of outer myometrial layer of myoma node (OR 3.06, p = .02), myoma size (OR 0.86, p = .04), and intramural extension degree (OR 0.91, p = .03) were significantly associated with outcome of complete resection. CONCLUSION: Thickness of outer myometrial layer of myoma node, myoma size, and intramural extension degree predicted outcome of 1-step hysteroscopic myomectomy. The chance of performing successful surgery increased with increased thickness of outer myometrial layer of myoma, and decreased with larger myomas and greater degrees of intramural extension.


Subject(s)
Hysteroscopy/methods , Leiomyoma/surgery , Uterine Neoplasms/surgery , Adult , Case-Control Studies , Female , Humans , Leiomyoma/pathology , Myometrium , Retrospective Studies , Surgical Instruments , Treatment Outcome , Uterine Neoplasms/pathology
3.
Fertil Steril ; 86(4 Suppl): 1146-51, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16962108

ABSTRACT

OBJECTIVE: To investigate the effects of a selective cyclooxygenase (COX)-2 inhibitor on endometriosis xenografts in immunodeficient mice. DESIGN: Prospective placebo-controlled study. SETTING: An academic facility at a Japanese university graduate school of medicine. PATIENT(S): Eight human ovarian endometriomas from seven patients. ANIMAL(S): Twenty-three female severe combined immunodeficiency (SCID) mice. INTERVENTION(S): Human ovarian endometriomas were implanted into the peritonea of SCID mice. Vehicle alone or NS398 (a selective COX-2 inhibitor, 10 mg/kg of weight per day) were administered orally daily for 56 days after implantation. Mice were killed on the 56th day. MAIN OUTCOME MEASURE(S): Change in explants size and immunohistochemical analyses to evaluating the proliferation index, apoptosis index, microvessel density, and labeling index assessing vascular endothelial growth factor and COX-2 expression by the endometriotic lesion. RESULT(S): NS398 significantly decreased implant size in comparison to vehicle alone (NS398 [medians, with range in brackets]: 22.0% [19.0%-36.7%] vs. vehicle: 41.2% [31.0%-55.3%], P<.01). Microvessel density (85.3 per mm2 [53.9-157.0 per mm2] vs. 121.8 per mm2 [97.2-259.6 per mm2], P=.02) and the vascular endothelial growth factor (0.4 [0-1.1] vs. 0.6 [0.5-2.1], P=.03) and COX-2 (0.4 [0.4-0.5] vs. 0.6 [0.4-0.8], P=.03) labeling indices in stromal cells were significantly lower in the NS398 group than in the vehicle group. There were no differences in the proliferation or apoptosis indices between the two groups. CONCLUSION(S): Selective COX-2 inhibitors decreased the size of implants and effectively treated endometriosis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Endometriosis/pathology , Endometriosis/physiopathology , Mice, SCID , Neovascularization, Pathologic/physiopathology , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , Adult , Animals , Apoptosis , Blood Vessels/pathology , Cell Proliferation/drug effects , Endometriosis/metabolism , Female , Humans , Immunohistochemistry , Immunologic Techniques , Mice , Microcirculation/drug effects , Middle Aged , Neovascularization, Pathologic/pathology , Peritoneum/surgery , Staining and Labeling , Transplantation, Heterologous
4.
Clin Cancer Res ; 12(14 Pt 1): 4200-8, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16857792

ABSTRACT

PURPOSE: In this study, we evaluated the correlation between endometrial carcinoma and peroxisome proliferator-activated receptor gamma (PPARgamma) expression and assessed whether PPARgamma ligands influence carcinoma growth. EXPERIMENTAL DESIGN: We examined the presence and cellular distribution of PPARgamma protein in 42 normal endometria, 32 endometria with hyperplasia, and 103 endometria with endometrial carcinoma by immunohistochemistry. We then compared PPARgamma mRNA expression in endometrial carcinoma with that in normal endometria using real-time reverse transcription-PCR. We subsequently confirmed expression of PPARgamma mRNA by real-time reverse transcription-PCR and PPARgamma protein by immunoblotting in endometrial carcinoma cell lines (Ishikawa, Sawano, and RL95-2 cells). We further examined the effects of PPARgamma agonist 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), a naturally occurring PPARgamma ligand, to these endometrial carcinoma cell lines. We also examined the status of apoptosis and p21 mRNA expression of these endometrial carcinoma cell lines following addition of 15d-PGJ2. RESULTS: PPARgamma immunoreactivity was detected in 11 of 23 (48%) of proliferative-phase endometrium, 14 of 19 (74%) of secretory-phase endometrium, 27 of 32 (84%) of endometrial hyperplasia, and 67 of 103 (65%) of carcinoma cases. PPARgamma immunoreactivity was significantly lower in endometrial carcinoma than in secretory-phase endometrium (P = 0.012) and endometrial hyperplasia (P = 0.006). There was a significant positive association between the status of PPARgamma and p21 expression in endometrial carcinoma (P < 0.0001). There was a significant negative association between the body mass index and PPARgamma labeling index of carcinoma tissue in the patients with endometrial carcinoma (P < 0.0001). PPARgamma mRNA was expressed abundantly in normal endometria but not in endometrial carcinoma. We showed that PPARgamma agonist 15d-PGJ2 inhibited cell proliferation and induced p21 mRNA of endometrial carcinoma cell lines. CONCLUSION: We showed the expression of PPARgamma in human endometrial carcinoma and the effects of PPARgamma ligand in endometrial carcinoma cells. These findings suggest that a PPARgamma ligand, 15d-PGJ2, has antiproliferative activity against endometrial carcinoma.


Subject(s)
Endometrial Neoplasms/pathology , PPAR gamma/physiology , Uterine Neoplasms/pathology , Aged , Cell Line, Tumor , Cell Proliferation , Endometrial Neoplasms/metabolism , Female , Humans , Ligands , Middle Aged , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Uterine Neoplasms/metabolism
6.
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
7.
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
8.
J Obstet Gynaecol Res ; 31(3): 216-23, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15916657

ABSTRACT

Hysteroscopic myomectomy is regarded as the best treatment for patients with submucous myomata. However, this procedure has a number of associated complications, including uterine perforation, cervical laceration, hyponatremia and hemorrhage, especially in cases of sessile submucous myomata. To avoid these problems, it is important to make well-advised preparations and manipulations both pre- and intraoperatively. The main surgical considerations for safe hysteroscopic myomectomy are shortening the operating time and avoiding cutting too deeply into the myometrium. With these requirements in mind, a combination of techniques using vaporization and a powerful oxytocic agent, such as prostaglandin F-2alpha, appears to be the safest method of carrying out hysteroresectoscopy for unpedunculated sessile submucous myomata.


Subject(s)
Hysteroscopy/methods , Leiomyoma/surgery , Uterine Neoplasms/surgery , Female , Humans , Hysteroscopy/adverse effects , Postoperative Complications
9.
Fertil Steril ; 82(3): 673-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15374713

ABSTRACT

OBJECTIVE: To determine the role of P and its nuclear receptor PR in the growth of ectopic uterine tissue of mice with or without a disrupted PR gene. DESIGN: Animal study. SETTING: Academic medical center. ANIMAL(S): Female wild-type (WT) and transgenic knockout mice for P receptor (PRKO). INTERVENTION(S): Endometriosis was induced in the following groups of ovariectomized adult mice: [1] untreated WT, [2] estradiol (E(2))-treated WT, [3] P-treated WT, [4] E(2) + P-treated WT, [5] untreated PRKO, [6] E(2)-treated PRKO, and [7] E(2) + P-treated PRKO (n = 5 in each group). MAIN OUTCOME MEASURE(S): The size of ectopic uterine tissue in WT and PRKO mice were compared between the groups subjected to treatments with P or E(2). Tissue proliferating cell nuclear antigen (PCNA) levels were compared among these groups. RESULTS: Treatment with P only significantly decreased the size of WT ectopic uterine tissue. The untreated PRKO ectopic uterine tissue was significantly larger than WT tissue. Estradiol increased the size of ectopic uterine tissues, and this E(2)-dependent growth could be suppressed by P in WT tissues but not in PRKO tissues. Finally, the hormone-dependent changes in ectopic uterine tissue size were accompanied by comparable alterations in PCNA levels. CONCLUSION(S): Intact PR in ectopic uterine tissue is essential to abolish E(2)-dependent or -independent proliferation. We also suggest that ectopic uterine tissue is associated with significantly increased resistance to P action and increased predisposition to E(2)-dependent proliferation in the absence of PR. Overall, these findings support the hypothesis that P resistance in human endometriosis may be due to the absence of sufficient levels of functional PR in this tissue.


Subject(s)
Cell Division/physiology , Endometriosis/pathology , Estradiol/pharmacology , Receptors, Progesterone/physiology , Animals , Cell Division/drug effects , Disease Models, Animal , Endometriosis/genetics , Female , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics
10.
Fertil Steril ; 81(5): 1351-6, 2004 May.
Article in English | MEDLINE | ID: mdl-15136101

ABSTRACT

OBJECTIVE: To investigate the effects of 17beta-estradiol (E(2)) on cyclooxygenase-2 (COX-2) expression and prostaglandin E(2) (PGE(2)) synthesis in primary human uterine microvascular endothelial cells (HUMEC). DESIGN: Prospective study. SETTING: Basic research laboratory at an academic medical center. PATIENT(S): Primary HUMEC of three women donors and primary human dermal microvascular endothelial cells of three women donors (as control), purchased from a third-party source. INTERVENTION(S): The HUMEC were cultured in specific media in a humidified atmosphere with 5% CO(2) at 37 degrees C. MAIN OUTCOME MEASURE(S): Measures of COX-2 mRNA and protein, PGE(2) production, and estrogen receptor alpha and beta mRNA and protein. RESULT(S): Treatment with E(2) (10(-10) to 10(-6) M) increased COX-2 mRNA levels by 2.3-fold to 2.4-fold in HUMEC. Treatment of HUMEC with E(2) (10(-8) M) resulted in a time-dependent increase of COX-2 mRNA levels. This was accompanied by a 2.8-fold increase in COX-2 protein level and a 1.5-fold increase in PGE(2) synthesis. Pretreatment of HUMEC with a selective COX-2 inhibitor, NS-398, abolished E(2)-induced PGE(2) synthesis, suggesting that E(2) specifically up-regulates COX-2 activity. The estrogen receptor antagonist ICI 182,780 fully reversed the stimulation of COX-2 mRNA and protein levels and PGE(2) synthesis by E(2). Interestingly, estrogen receptor beta mRNA and protein were abundant in HUMEC, whereas estrogen receptor alpha mRNA or protein was barely detectable. CONCLUSION(S): We conclude that various levels of E(2) can significantly increase COX-2 expression and PGE(2) synthesis in HUMEC via the estrogen receptor.


Subject(s)
Endothelial Cells/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Estrogen/physiology , Uterus/drug effects , Cells, Cultured , Cyclooxygenase 2 , Dinoprostone/biosynthesis , Endothelial Cells/enzymology , Female , Fulvestrant , Humans , Isoenzymes/analysis , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/analysis , Up-Regulation , Uterus/blood supply , Uterus/enzymology
11.
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
12.
Mol Cell Endocrinol ; 208(1-2): 61-75, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-14580722

ABSTRACT

Binding activity of steroidogenic factor-1 (SF-1) to promoters of the majority of steroidogenic genes in response to gonadotropins is a critical mechanism that regulates steroidogenesis in gonads. Thus, the modulation of SF-1 action may be essential for the differential regulation of formation of sex steroids in the ovary. Aromatase P450 (P450arom) is the rate-limiting enzyme for estrogen formation. In this study, we characterize another nuclear receptor half site in the gonadal aromatase promoter which we show to be important for aromatase regulation. We also show herein that the stimulation of P450arom promoter activity by SF-1 in ovarian granulosa, testicular Sertoli and JEG-3 choriocarcinoma cells is inhibited by two transcription factors, Wilms' tumor suppressor gene (WT1) and dosage sensitive sex reversal adrenal hypoplasia congenita critical region on the X chromosome gene 1 (DAX-1). Given the characterized roles of these transcription factors in gonadal development and function, modulation of SF-1 action by WT1 and DAX-1 may represent an important key mechanism in steroidogenesis.


Subject(s)
Aromatase/genetics , DNA-Binding Proteins/physiology , Gonads/metabolism , Receptors, Retinoic Acid/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , WT1 Proteins/physiology , Amino Acid Motifs , Animals , Aromatase/metabolism , Base Sequence , Cell Line , Consensus Sequence , DAX-1 Orphan Nuclear Receptor , Down-Regulation , Female , Fushi Tarazu Transcription Factors , Gene Expression Regulation , Genes, Wilms Tumor , Humans , Male , Mice , Molecular Sequence Data , Plasmids , Promoter Regions, Genetic , Sertoli Cells/metabolism , Steroidogenic Factor 1 , Transcriptional Activation
13.
J Clin Endocrinol Metab ; 87(9): 4369-77, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213901

ABSTRACT

The orphan nuclear receptor steroidogenic factor-1 (SF-1) induces the expression of Müllerian inhibiting substance (MIS) and many steroidogenic genes, including aromatase P450 (P450arom). Dosage-sensitive sex reversal adrenal hypoplasia congenita critical region on the X chromosome gene 1 (DAX-1) inhibits SF-1-mediated induction of MIS and other steroidogenic genes, whereas Wilms' tumor suppressor gene (WT1) augments SF-1-mediated MIS expression. The effects of WT1 on steroidogenesis or P450arom expression have not been explored to date. In human endometriotic stromal cells, extremely high levels of P450arom mRNA and enzyme activity are present. Prostaglandin E(2) stimulates cAMP formation, SF-1 binding activity, P450arom mRNA levels, and estrogen synthesis in endometriotic stromal cells. Stromal cells of eutopic endometrium from disease-free women, on other hand, do not contain readily detectable levels of P450arom mRNA. Thus, we evaluated herein the possible roles of WT1 and DAX-1 in cAMP/SF-1-mediated regulation of P450arom expression in endometriotic and endometrial stromal cells. We also determined the cellular distribution and levels of these transcription factors in pathological endometriotic vs. normal eutopic endometrial tissues by immunohistochemistry to understand their in vivo roles. In vitro transcriptional regulation studies showed that both WT1 and DAX-1 inhibited cAMP and/or SF-1-induced P450arom promoter activity in a dose-dependent fashion in cultured human endometriotic and endometrial stromal cells. Site-directed disruption of the SF-1 binding site (-136/-124 bp) in the P450arom promoter abolished basal or cAMP/SF-1-induced promoter activity in the presence or absence of WT1 or DAX-1. Immunohistochemistry and H-scoring showed that DAX-1 was ubiquitously present in epithelial and stromal cells of both tissues. WT1, on the other hand, was preferentially expressed in stromal (vs. epithelial) cells. Moreover, WT1 levels in endometriotic stromal cells are significantly down-regulated compared with normal endometrial stromal cells. In summary, WT1 or DAX-1 inhibits cAMP-SF-1 pathway-dependent P450arom expression in cultured human endometriotic and endometrial stromal cells. In vivo down-regulation of WT1 in endometriotic stromal cells (vs. normal endometrial stromal cells) may in part be responsible for aberrantly increased P450arom expression and estrogen formation in this pathological tissue.


Subject(s)
Aromatase/genetics , DNA-Binding Proteins/genetics , Endometriosis/enzymology , Endometrium/enzymology , Gene Expression Regulation, Enzymologic/physiology , Receptors, Retinoic Acid/genetics , Repressor Proteins , Stromal Cells/enzymology , Transcription Factors/genetics , WT1 Proteins/genetics , 5' Untranslated Regions/genetics , Base Sequence , Biopsy , DAX-1 Orphan Nuclear Receptor , Endometriosis/pathology , Epithelial Cells/enzymology , Epithelial Cells/pathology , Female , Humans , Luciferases/genetics , Luciferases/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Steroidogenic Factor 1
14.
J Clin Endocrinol Metab ; 87(7): 3263-73, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12107235

ABSTRACT

We investigated the regulation of PG production in human endometrial stromal cells (ESC) by IL-1beta. We found that cyclooxygenase-2 (COX-2) mRNA and protein levels and PGE(2) production in ESC were significantly increased by IL-1beta. COX-2 mRNA, protein, and PGE(2) levels in IL-1beta-treated ESC were decreased by a PKA inhibitor, a nuclear factor (NF-kappaB) inhibitor, and an ERK1/2 inhibitor, but not by a p38 MAPK inhibitor or a PKC inhibitor, suggesting the possible involvement of PKA, NF-kappaB, and/or the ERK1/2 signaling pathway(s) in IL-1beta-mediated COX-2 gene induction in ESC. We then transiently transfected deletion mutants of the COX-2 promoter fused to the luciferase reporter gene and variants of -360/+56 bp promoter construct carrying different site-directed mutations of selected cis-acting elements. We determined that a NF-kappaB site (-222/-213 bp), a nuclear factor for IL-6 expression site (NF-IL6, -132/-124 bp), and a cAMP response element (-59/-52 bp) were essential for the baseline COX-2 gene promoter regulation. The addition of IL-1beta, however, did not affect the activity of these COX-2 promoter constructs. To investigate the potential effects of IL-1beta on COX-2 mRNA stability, ESC were treated with actinomycin D, a general transcription inhibitor, in the absence or presence of IL-1beta. We found that 1) IL-1beta significantly increased COX-2 mRNA stability; 2) continuous transcription was not required to sustain the IL-1beta-induced COX-2 mRNA levels; and 3) COX-2 mRNA was highly unstable in the absence of IL-1beta. Additionally, we found that the ERK1/2 signaling pathway was essential for stabilizing COX-2 mRNA. We conclude that levels of COX-2 mRNA, protein, and enzyme activity in ESC are controlled by various signaling pathways, including PKA, ERK1/2, and NF-kappaB. Moreover, posttranscriptional mRNA stability is an important mechanism for IL-1beta-induced elevation of COX-2 expression in ESC.


Subject(s)
Endometrium/metabolism , Interleukin-1/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Proline/analogs & derivatives , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/metabolism , Stromal Cells/metabolism , Butadienes/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclooxygenase 2 , Dinoprostone/biosynthesis , Endometrium/cytology , Enzyme Inhibitors/pharmacology , Extracellular Space/metabolism , Female , Humans , Membrane Proteins , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Proline/pharmacology , Promoter Regions, Genetic/physiology , RNA Stability/drug effects , Reference Values , Signal Transduction , Stromal Cells/drug effects , Thiocarbamates/pharmacology
15.
J Clin Endocrinol Metab ; 87(7): 3460-6, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12107266

ABSTRACT

Aromatase P450 (P450arom) is the key enzyme for the biosynthesis of estrogen that is essential for the growth of human endometriosis, a pathology characterized by endometrium-like tissue on the peritoneal surfaces of abdominal organs manifest by pelvic pain and infertility. Surgically transplanted autologous uterine tissue to ectopic sites on the peritoneum in mice has been used as an animal model to study endometriosis. Using this mouse model, we evaluated the roles of the P450arom gene and aromatase enzyme activity in the growth of endometriosis represented by ectopic uterine tissues in mice. Endometriosis was induced surgically in the following groups of mice: 1) untreated transgenic mice with disrupted P450arom gene (ArKO); 2) ArKO mice treated with systemic estrogen; 3) untreated wild-type (WT) mice; 4) WT mice treated with estrogen; 5) WT mice treated with the aromatase inhibitor, letrozole; and 6) WT mice treated with letrozole and estrogen. Each group contained eight mice; +/+ littermates of ArKO mice were used as WT controls. Treatment with estrogen increased the size of ectopic uterine tissues in ArKO and WT mice significantly. The ectopic uterine lesions in untreated and estrogen-treated ArKO mice were strikingly smaller than those in untreated and estrogen-treated WT controls, respectively. Systemic treatment of WT mice with letrozole significantly decreased the lesion size in a dose-dependent manner. The addition of estrogen to letrozole treatment increased the ectopic lesion size, although these lesions were significantly smaller than those in mice treated with estrogen only. As tissue controls, the effects of these conditions on normally located (eutopic) uterine tissue were evaluated. The effects of disruption of the P450arom gene and treatments with letrozole and estrogen seemed to be more profound on ectopic tissues, suggesting that ectopic tissues might be more sensitive to estrogen for growth. We conclude that both an intact P450arom gene and the presence of aromatase enzyme activity are essential for the growth of ectopic uterine tissue in a mouse model of endometriosis.


Subject(s)
Aromatase/physiology , Endometriosis/physiopathology , Animals , Aromatase/genetics , Aromatase Inhibitors , Cell Division/physiology , Choristoma/pathology , Choristoma/physiopathology , Endometriosis/pathology , Enzyme Inhibitors/pharmacology , Estrogens/pharmacology , Female , Growth/drug effects , Letrozole , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Nitriles/pharmacology , Triazoles/pharmacology , Uterus/drug effects , Uterus/pathology
16.
J Clin Endocrinol Metab ; 87(7): 3504-7, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12107271

ABSTRACT

We investigated the effects of vascular endothelial growth factor (VEGF) on cyclooxygenase-2 (COX-2) expression and prostaglandin E(2) (PGE(2)) synthesis in human microvascular endothelial cells (HMEC-1). Treatment of HMEC-1 with VEGF resulted in a dose- and time-dependent up-regulation of COX-2 mRNA and protein levels. This up-regulation was accompanied by a 1.6-fold increase in PGE(2) synthesis. Pretreatment of HMEC-1 with a selective COX-2 inhibitor, NS-398, abolished VEGF-induced PGE(2) synthesis, suggesting specific up-regulation of COX-2 activity by VEGF in HMEC-1. Transient transfection assays using deletion mutants of the human COX-2 promoter fused to the luciferase reporter gene indicated critical requirement of a regulatory region spanning -828/-123 bp for VEGF induction of COX-2 promoter activity in HMEC-1. Site-directed mutation analysis demonstrated that a GATA cis-acting element at -685/-680 bp was essential for VEGF- induced COX-2 promoter activity in HMEC-1. These observations are of particular importance given the recent demonstrations of critical requirement of COX-2 isoenzyme for tumor growth and angiogenesis.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/metabolism , Isoenzymes/metabolism , Lymphokines/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Cell Line , Cyclooxygenase 2 , Dinoprostone/biosynthesis , Endothelium, Vascular/cytology , Humans , Isoenzymes/genetics , Membrane Proteins , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
J Reprod Immunol ; 55(1-2): 21-33, 2002.
Article in English | MEDLINE | ID: mdl-12062819

ABSTRACT

Estrogen is produced in a number of human tissues including the ovary, placenta and extraglandular sites such as adipose tissue, skin and the brain. Aromatase is the key enzyme that regulates estrogen formation in these tissues. Aromatase activity is not detectable in normal endometrium. In contrast, aromatase is expressed aberrantly in endometriosis and is stimulated by PGE(2). This results in local production of estrogen, which induces PGE(2) formation and establishes a positive feedback cycle. Another abnormality in endometriosis, i.e. deficient 17beta-hydroxysteroid dehydrogenase (17beta-HSD) type 2 expression, impairs the inactivation of estradiol to estrone. These molecular aberrations collectively favor accumulation of increasing quantities of estradiol and PGE(2) in endometriosis. The clinical relevance of these findings was exemplified by the successful treatment of an unusually aggressive case of postmenopausal endometriosis using an aromatase inhibitor.


Subject(s)
Endometriosis/metabolism , Estrogens/biosynthesis , Aromatase/genetics , Aromatase/metabolism , Aromatase Inhibitors , Endometriosis/drug therapy , Endometriosis/etiology , Enzyme Inhibitors/therapeutic use , Estradiol/metabolism , Female , Gene Expression , Humans , Inflammation/etiology , Inflammation/metabolism , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
18.
J Biol Chem ; 277(29): 26208-16, 2002 Jul 19.
Article in English | MEDLINE | ID: mdl-12006564

ABSTRACT

We investigated the regulation of prostaglandin production in normal endometrial stromal cells (ESC) by malignant endometrial epithelial cells. We found that cyclooxygenase (COX)-2 mRNA and protein levels and prostaglandin (PG)E(2) production in ESC were significantly increased by Ishikawa malignant endometrial epithelial cell conditioned medium (MECM). By using transient transfection assays, we found that the -360/-218-bp region of the COX-2 promoter gene was critical for MECM induction of promoter activity. This MECM-responsive region contained a variant nuclear factor (NF)-kappa B site at -222 to -213 that, when mutated, completely abolished COX-2 promoter activation by MECM. Employing electrophoretic mobility shift assays, we further demonstrated that binding of NF-kappa B p65 to this NF-kappa B-binding site is, in part, responsible for the COX-2 promoter activation by MECM. To investigate further the potential effects of MECM on COX-2 mRNA stability, ESC were treated with MECM in the absence or presence of actinomycin D, a general transcription inhibitor. We found that MECM significantly increased COX-2 mRNA stability. Intriguingly, we found that PGE(2) was one of the major factors in MECM, which was responsible for up-regulating COX-2 expression in ESC. ECC-1 and HEC-1A malignant endometrial epithelial cell lines also produced significantly increased quantities of PGE(2). In conclusion, malignant endometrial epithelial cells secrete PGE(2) that induces COX-2 expression in normal endometrial stromal cells in a paracrine fashion through activation of transcription and stabilization of COX-2 mRNA.


Subject(s)
Dinoprostone/metabolism , Endometrial Neoplasms/enzymology , Endometrium/enzymology , Gene Expression Regulation, Enzymologic , Isoenzymes/biosynthesis , NF-kappa B/metabolism , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandins/biosynthesis , Up-Regulation , Cyclooxygenase 2 , Epithelial Cells/enzymology , Female , Humans , Isoenzymes/genetics , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/genetics , Stromal Cells/enzymology , Tumor Cells, Cultured
19.
J Clin Endocrinol Metab ; 87(5): 2336-45, 2002 May.
Article in English | MEDLINE | ID: mdl-11994385

ABSTRACT

In human endometriotic stromal cells, markedly high levels of aromatase P450 (P450arom) mRNA and promoter II activity are present and can be vigorously stimulated by PGE(2) via a cAMP-dependent pathway to give rise to physiologically significant estrogen biosynthesis. Stromal cells of eutopic endometrium, on the other hand, do not express sufficient levels of P450arom for detectable enzyme activity. Because P450arom is up-regulated in the ovaries of CCAAT/enhancer binding protein (C/EBP) beta knockout mice and activation of the ovarian-type P450arom promoter (II) is responsible for aberrant P450arom expression in endometriosis, we sought here to evaluate the possible roles of C/EBP isoforms in the regulation of P450arom expression in endometriotic vs. eutopic endometrial stromal cells. We previously found that the -517-bp flanking region of promoter II contained the critical cis-acting elements for baseline and cAMP (analog)-induced activity. In this study, we disrupted several potential sequences and found that mutations of a -211/-197-bp cAMP-response element (CRE) and a -317/-304-bp C/EBP binding site abolished both baseline and cAMP-induced promoter II activity. Ectopic expression of C/EBPalpha increased both baseline and cAMP-dependent promoter II activity significantly in endometriotic cells, whereas ectopic expression of C/EBPbeta or C/EBPdelta abolished promoter II activity in both untreated and cAMP-treated endometriotic stromal cells. Comparable changes in promoter II activity were observed using endometrial stromal cells, which showed, however, seemingly diminished levels of baseline and cAMP-induced promoter II activity in comparison with endometriotic cells. EMSA using a probe containing the critical -317/-304-bp C/EBP site upstream of promoter II demonstrated a distinct DNA-protein complex in endometriotic, but not in endometrial stromal cells. This specific complex, however, could not be altered using antibodies against C/EBPalpha, -beta, or -delta. Because CRE is another potential DNA motif that can bind C/EBP isoforms, we next used EMSA using a probe containing the -211/-197-bp CRE and demonstrated that specific DNA-protein complexes contained C/EBPalpha but not C/EBPbeta or C/EBPdelta in endometriotic stromal cells. In contrast, C/EBPbeta and C/EBPdelta but not C/EBPalpha were detected in DNA-protein complexes using nuclear extracts from endometrial stromal cells. Western blotting and immunohistochemistry demonstrated expression of C/EBPalpha, -beta, and -delta in human endometriotic and endometrial stroma and epithelium. Intriguingly, C/EBPbeta was expressed at increased levels in stromal cells of human eutopic endometrium compared with simultaneously biopsied endometriotic tissues. We conclude that both -317/-304 and -211/-197-bp elements in promoter II are critical for the robust cAMP-dependent induction in endometriosis. C/EBPalpha up-regulates, whereas C/EBPbeta and C/EBPdelta inhibit P450arom promoter activity via binding primarily to the -211/-197-bp CRE under in vitro conditions. In vivo down-regulation of C/EBPbeta in endometriotic stromal cells and its up-regulation in endometrial stromal cells may in part account for the induction of P450arom expression in endometriosis and its inhibition in endometrium.


Subject(s)
Aromatase/genetics , CCAAT-Enhancer-Binding Proteins/physiology , Endometriosis/genetics , Endometrium/physiology , Gene Expression Regulation/physiology , Stromal Cells/physiology , Adult , Aromatase/metabolism , CCAAT-Enhancer-Binding Protein-alpha/physiology , CCAAT-Enhancer-Binding Protein-beta/physiology , Cells, Cultured , Cyclic AMP/physiology , Endometriosis/pathology , Endometrium/cytology , Female , Humans , Nuclear Proteins/metabolism , Promoter Regions, Genetic/physiology , Response Elements/physiology , Tissue Distribution , Transcription, Genetic/physiology
20.
Ann N Y Acad Sci ; 955: 75-85; discussion 86-8, 396-406, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11949967

ABSTRACT

Aromatase activity is absent in normal endometrium. In contrast, aromatase is expressed aberrantly in endometriosis, which gives rise to strikingly high levels of aromatase activity in this tissue. Both aromatase expression and activity are stimulated by PGE2. This results in local production of estrogen, which induces PGE2 formation and establishes a positive feedback cycle. Another abnormality in endometriosis, that is, deficient 17beta-HSD type 2 expression, impairs the inactivation of estradiol to estrone. These molecular aberrations collectively favor accumulation of increasing quantities of estradiol and PGE2 in endometriosis. The clinical relevance of these findings was exemplified by the successful treatment of an unusually aggressive case of postmenopausal endometriosis using an aromatase inhibitor.


Subject(s)
Endometriosis/metabolism , Estrogens/biosynthesis , Aromatase/metabolism , Aromatase Inhibitors , Dinoprostone/metabolism , Endometriosis/drug therapy , Endometriosis/enzymology , Endometrium/enzymology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Estrogens/metabolism , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...