Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Steroid Biochem Mol Biol ; 112(1-3): 102-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18832036

ABSTRACT

We characterised the effects of selective oestrogen receptor modulators (SERM) in explant cultures of human endometrium tissue. Endometrium tissues were cultured for 24h in Millicell-CM culture inserts in serum-free medium in the presence of vehicle, 17beta-estradiol (17beta-E2, 1nM), oestrogen receptor (ER) antagonist ICI 164.384 (40nM), and 4-OH-tamoxifen (40nM), raloxifene (4nM), lasofoxifene (4nM) and acolbifene (4nM). Protein expression of ERalpha, ERbeta1 and Ki-67 were evaluated by immunohistochemistry (IHC). The proliferative fraction was assessed by counting the number of Ki-67 positive cells. Nuclear staining of ER( and ER(1 was observed in the glandular epithelium and stroma of pre- and postmenopausal endometrium. ER(1 protein was also localized in the endothelial cells of blood vessels. Treating premenopausal endometrium tissue with 17beta-E2 increased the fraction of Ki-67 positive cells (p<0.001) by 55% in glands compared to the control. Raloxifene (4nM) increased (p<0.05) the Ki-67 positive fraction. All other SERMS did not affect proliferation in this model. Treating postmenopausal endometrium with 17(-E2 increased (p<0.001) the fraction of Ki-67 positive cells by 250% in glands compared to the control. A similar effect was also seen for 4-OH-tamoxifen, whereas the rest of SERMs did not stimulate proliferation. We demonstrated that oestradiol increases the fraction of proliferating cells in short term explant cultures of postmenopausal endometrium. In addition, we were able to reveal the agonistic properties of 4-OH-tamoxifen and confirm that raloxifene and next-generation SERMs acolbifene and lasofoxifene were neutral on the human postmenopausal endometrium.


Subject(s)
Cell Proliferation/drug effects , Endometrium/drug effects , Postmenopause , Premenopause , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Adult , Cells, Cultured , Endometrium/cytology , Female , Humans , Middle Aged , Selective Estrogen Receptor Modulators/metabolism
2.
Hum Reprod ; 23(12): 2692-700, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18716038

ABSTRACT

BACKGROUND: In this study, we characterized the fibromuscular (FM) tissue, typical of deeply infiltrating endometriosis, investigated which cells are responsible for the FM reaction and evaluated whether transforming growth factor-beta (TGF-beta) signaling is involved in this process. METHODS: FM differentiation and TGF-beta signaling were assessed in deeply infiltrating endometriosis lesions (n = 20) and a nude mouse model of endometriosis 1, 2, 3 and 4 weeks post-transplantation. The FM reaction was evaluated by immunohistochemistry using different markers of FM and smooth muscle cell differentiation (vimentin, desmin, alpha-smooth muscle actin, smooth muscle myosin heavy chain). TGF-beta signaling was assessed by immunostaining for its receptors and phosphorylated Smad. RESULTS: Deeply infiltrating endometriosis lesions contain myofibroblast-like cells that express multiple markers of FM differentiation. Expression of TGF-beta receptors and phospho-Smad was more pronounced in the endometrial component of the lesions than in the FM component. In the nude mouse model, alpha-smooth muscle actin expression was observed in murine fibroblasts surrounding the lesion, but not in human endometrial stroma. CONCLUSIONS: FM differentiation in deeply infiltrating endometriosis is the result of a reaction of the local environment to the presence of ectopic endometrium. It shares characteristics with pathological wound healing, but cannot be explained by TGF-beta signaling alone.


Subject(s)
Endometriosis/pathology , Animals , Cell Differentiation , Choristoma/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Mice, Nude , Muscle, Smooth/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism
3.
Hum Reprod ; 23(3): 635-41, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18216035

ABSTRACT

BACKGROUND: The general concept that haemoglobin is only a carrier protein for oxygen and carbon dioxide is challenged since recent studies have shown haemoglobin expression in non-erythroid cells and the protection of haemoglobin against oxidative and nitrosative stress. Using microarrays, we previously showed expression of haemoglobins alpha, beta, delta and gamma and the haeme metabolizing enzyme, haeme oxygenase (HO)-1 in human endometrium. METHODS: Using real-time quantitative PCR, haemoglobin alpha, beta, delta and gamma, and HO-1 mRNA levels were assessed throughout the menstrual cycle (n = 30 women). Haemoglobin and HO-1 protein levels in the human endometrium were assessed with immunohistochemistry. For steroid responsiveness, menstrual and late proliferative-phase endometrial explants were cultured for 24 h in the presence of vehicle (0.1% ethanol), estradiol (17beta-E(2,) 1 nM), progestin (Org 2058, 1 nM) or 17beta-E(2)+Org 2058 (1 nM each). RESULTS: All haemoglobins and the HO-1 were expressed in normal human endometrium. Haemoglobin mRNA and protein expression did not vary significantly during the menstrual cycle. Explant culture with Org 2058 or 17beta-E(2)+Org 2058 increased haemoglobin gamma mRNA expression (P < 0.05). HO-1 mRNA levels, and not protein levels, were significantly higher during the menstrual (M)-phase of the cycle (P < 0.05), and were down-regulated by Org 2058 in M-phase explants and by 17beta-E(2)+Org 2058 in LP-phase explants, versus control (P < 0.05). CONCLUSIONS: The haemoglobin-HO-1 system may be required to ensure adequate regulation of the bioavailability of haeme, iron and oxygen in human endometrium.


Subject(s)
Endometrium/metabolism , Hemoglobins/biosynthesis , Adult , Endometrium/drug effects , Estradiol/pharmacology , Female , Heme Oxygenase-1/biosynthesis , Humans , Immunohistochemistry , Menstrual Cycle , Middle Aged , Polymerase Chain Reaction , Pregnenediones/pharmacology , RNA, Messenger/metabolism , Tissue Culture Techniques
4.
Mol Hum Reprod ; 13(12): 875-86, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17989082

ABSTRACT

To identify specific markers of rectovaginal endometriotic nodule vasculature, highly enriched preparations of vascular endothelial cells and pericytes were obtained from endometriotic nodules and control endometrial and myometrial tissue by laser capture microdissection (LCM), and gene expression profiles were screened by microarray analysis. Of the 18 400 transcripts on the arrays, 734 were significantly overexpressed in vessels from fibromuscular tissue and 923 in vessels from stromal tissue of endometriotic nodules, compared with vessels dissected from control tissues. The most frequently expressed transcripts included known endothelial cell-associated genes, as well as transcripts with little or no previous association with vascular cells. The higher expression in blood vessels was further corroborated by immunohistochemical staining of six potential markers, five of which showed strong expression in pericytes. The most promising marker was matrix Gla protein, which was found to be present in both glandular epithelial cells and vascular endothelial cells of endometriotic lesions, although it was barely expressed at all in normal endometrium. LCM, combined with microarray analysis, constitutes a powerful tool for mapping the transcriptome of vascular cells. After immunohistochemical validation, markers of vascular endothelial and perivascular cells from endometriotic nodules could be identified, which may provide targets to improve early diagnosis or to selectively deliver therapeutic agents.


Subject(s)
Antigens/immunology , Blood Vessels/immunology , Endometriosis/immunology , Adult , Antigens/genetics , Antigens/metabolism , Blood Vessels/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Endometriosis/genetics , Endometriosis/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Humans , Immunohistochemistry , Microdissection , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Matrix Gla Protein
5.
Hum Reprod ; 22(12): 3148-58, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17921479

ABSTRACT

BACKGROUND: Estradiol (E(2)) is an important promoter of the growth of both eutopic and ectopic endometrium. The findings with regard to the expression and activity of steroidogenic enzymes in endometrium of controls, in endometrium of endometriosis patients and in endometriotic lesions are not consistent. METHODS: In this study, we have looked at the mRNA expression and protein levels of a range of steroidogenic enzymes [aromatase, 17beta-hydroxysteroid dehydrogenases (17beta-HSD) type 1, 2 and 4, estrogen sulfotransferase (EST) and steroid sulfatase (STS)] in eutopic and ectopic endometrium of patients (n = 14) with deep-infiltrative endometriosis as well as in disease-free endometrium (n = 48) using real-time PCR and immunocytochemistry. In addition, we evaluated their menstrual cycle-related expression patterns, and investigated their steroid responsiveness in explant cultures. RESULTS: Aromatase and 17beta-HSD type 1 mRNA levels were extremely low in normal human endometrium, while mRNAs for types 2 and 4 17beta-HSD, EST and STS were readily detectable. Only 17beta-HSD type 2 and EST genes showed sensitivity to progesterone in normal endometrium. Types 1 and 2 17beta-HSD and STS protein was detected in normal endometrium using new polyclonal antibodies. CONCLUSIONS: In endometriosis lesions, the balance is tilted in favor of enzymes producing E(2). This is due to a suppression of types 2 and 4 17beta-HSD, and an increased expression of aromatase and type 1 17beta-HSD in ectopic endometrium.


Subject(s)
Endometriosis/metabolism , Endometrium/enzymology , Estrogens/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/immunology , 17-Hydroxysteroid Dehydrogenases/metabolism , Adult , Animals , Antibody Specificity , Aromatase/genetics , Aromatase/metabolism , Estrogens/biosynthesis , Female , Gene Expression Regulation, Enzymologic , Humans , Immunohistochemistry , Menstrual Cycle/metabolism , Middle Aged , RNA, Messenger/metabolism , Rabbits , Steryl-Sulfatase/genetics , Steryl-Sulfatase/immunology , Steryl-Sulfatase/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism , Tissue Culture Techniques
6.
Reprod Sci ; 14(4): 367-73, 2007 May.
Article in English | MEDLINE | ID: mdl-17644809

ABSTRACT

Deep infiltrating endometriosis is characterized by the presence of nodular lesions largely composed of fibromuscular tissue. Transforming growth factor beta 1 (TGF-beta1) is the cytokine most causatively associated with disorders characterized by fibrosis throughout the body. Therefore, the hypothesis was tested that mechanisms increasing the fraction of biologically active TGF-beta1, such as TGF-beta 1 gene polymorphisms, lead to an increased risk of developing deep infiltrating endometriosis. The frequency of the -509C/T polymorphism of the TGF-beta 1 gene was tested in women with deep infiltrating endometriosis (n = 72), gynecological patients without symptoms of endometriosis (n = 95) and healthy females (n = 93). Detection of the -509C/T polymorphisms was performed using PCR-restriction fragment length polymorphism analysis. We did not observe statistically significant differences in the frequency of the -509C/T polymorphism between the groups. Our study does not support an association between the -509C/T polymorphism of the TGF-beta 1 gene and an increased risk of deep infiltrating endometriosis.


Subject(s)
Endometriosis/genetics , Polymorphism, Single Nucleotide , Transforming Growth Factor beta1/genetics , Cytosine , DNA/genetics , DNA/isolation & purification , Endometriosis/pathology , Endometriosis/physiopathology , Endometriosis/surgery , Female , Genotype , Humans , Polymerase Chain Reaction , Thymine
7.
Hum Reprod Update ; 13(4): 405-17, 2007.
Article in English | MEDLINE | ID: mdl-17584823

ABSTRACT

To date, research into the biological processes and molecular mechanisms associated with endometrial receptivity and embryo implantation has been a focus of attention, whereas the complex events that occur in the human endometrium during the menstrual and proliferative phase under the influence of estrogen have received little attention. The objective of this review is to provide an update of our current understanding of the actions of estrogen on both human and rodent endometrium, with special emphasis on the regulation of uterine growth and cell proliferation, and the value of global gene expression analysis, in increasing understanding of these processes.


Subject(s)
Endometrium/metabolism , Estrogens/pharmacology , Gene Expression/drug effects , Animals , Cell Proliferation/drug effects , Endometrium/drug effects , Endometrium/growth & development , Estrogens/genetics , Estrogens/physiology , Female , Gene Expression Profiling , Genome, Human , Humans , Menstrual Cycle/drug effects , Menstrual Cycle/metabolism , Mice , Signal Transduction/drug effects , Signal Transduction/genetics , Uterus/drug effects , Uterus/growth & development , Uterus/metabolism
8.
J Steroid Biochem Mol Biol ; 104(3-5): 246-51, 2007 May.
Article in English | MEDLINE | ID: mdl-17481886

ABSTRACT

Disorders of estrogen-responsive tissues are frequently associated with aberrations in steroid metabolism due to altered expression of synthesizing and metabolizing enzymes. For instance, overexposure to unopposed 17beta-estradiol has been associated with the pathogenesis of endometrial proliferative disorders, such as endometriosis. Investigations into the metabolic conversion in tissues and cells have been rather limited. This is mostly due to fact that such studies have to make use of radioactive steroid hormones and expensive equipment to obtain sufficient sensitivity. We adapted a sensitive non-radioactive HPLC method to study estrogen metabolism in more detail. This HPLC method is based on the solid phase extraction of estrogens and the derivatization of the steroids with 2-(4-carboxy-phenyl)-5,6-dimethylbenzimidazole. The technique is sensitive, robust and is useful for the detection of aromatase, 17beta-HSD types 1 and 2 and sulfatase activities in lysates of placenta and endometrium.


Subject(s)
Chromatography, High Pressure Liquid/methods , Estrogens/analysis , Estrogens/metabolism , 17-Hydroxysteroid Dehydrogenases/analysis , Aromatase/analysis , Endometrium/enzymology , Endometrium/metabolism , Female , Humans , Models, Biological , Pilot Projects , Placenta/enzymology , Placenta/metabolism , Sensitivity and Specificity , Steryl-Sulfatase/analysis
9.
Cell Mol Life Sci ; 64(7-8): 1009-32, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17404688

ABSTRACT

Genomic profiling was performed on explants of late proliferative phase human endometrium after 24-h treatment with progesterone (P) or oestradiol and progesterone (17beta-E(2)+P) and on explants of menstrual phase endometrium treated with 17beta-E(2)+P. Gene expression was validated with real-time PCR in the samples used for the arrays, in endometrium collected from early and mid-secretory phase endometrium, and in additional experiments performed on new samples collected in the menstrual and late proliferative phase. The results show that late proliferative phase human endometrium is more responsive to progestins than menstrual phase endometrium, that the expression of several genes associated with embryo implantation (i.e. thrombomodulin, monoamine oxidase A, SPARC-like 1) can be induced by P in vitro, and that genes that are fully dependent on the continuous presence of 17beta-E(2) during P exposure can be distinguished from those that are P-dependent to a lesser extent. Therefore, 17beta-E(2) selectively primes implantation-related genes for the effects of P.


Subject(s)
Endometrium/metabolism , Estradiol/pharmacology , Estrogens/physiology , Gene Expression/drug effects , Progesterone/pharmacology , Adult , Embryo Implantation/genetics , Endometrium/drug effects , Female , Follicular Phase/metabolism , Humans , In Vitro Techniques , Menstrual Cycle/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
10.
Int J Gynecol Cancer ; 17(4): 897-904, 2007.
Article in English | MEDLINE | ID: mdl-17359291

ABSTRACT

Tumor hypoxia can trigger the induction of angiogenesis. High microvessel density (MVD) as well as hypoxia-inducible factor-1alpha (HIF-1alpha) have been related to recurrent disease and tumor aggressiveness, respectively. In this study, MVD and hypoxic status were investigated in primary and recurrent endometrial carcinomas. A total of 65 primary tumors of patients with recurrent endometrial carcinoma (n = 40), and without recurrent endometrial carcinoma (n = 25) were studied. Immunohistochemical analysis was performed on paraffin-embedded tumor tissue. MVD was determined by quantitative analysis of CD31/FVIII positive vessels. Tumor hypoxia was estimated by evaluating the expression of the hypoxia-regulated gene HIF-1alphaand its target gene carbonic anhydrase IX (CA-IX). An additional 23 recurrent tumors were available for determination of MVD and HIF-1alpha expression. Effects of hypoxia on tumor protein p53 (TP53) expression were evaluated in the endometrial cancer cell lines (ECC-1), Ishikawa (derived from adenocarcinomas), and AN3CA (derived from a lymph node metastasis). MVD, CA-IX, and HIF-1alpha expression were not significantly different in primary tumors of patients with recurrence compared to the control tumors. The MVD was significantly lower, and HIF-1alpha expression was significantly higher in recurrent tumors when compared with their primary tumors (paired t test, P < 0.05). HIF-1alpha expression correlated well with TP53 expression levels in primary tumors, but not in recurrences. TP53 protein levels were highest in AN3CA cells. Hypoxic conditions induced TP53 protein in ECC-1 and Ishikawa, but not AN3CA cells. We conclude that MVD, CA-IX, and HIF-1alpha expression are not independent prognostic markers for recurrent endometrial carcinoma. The low MVD, increased HIF-1alpha protein levels, dissociation of hypoxia, and TP53 protein induction in the metastatic tumor cells (AN3CA) support a role for hypoxia in the development of recurrent endometrial carcinoma.


Subject(s)
Carcinoma, Endometrioid/blood supply , Endometrial Neoplasms/blood supply , Endometrial Neoplasms/metabolism , Neoplasm Recurrence, Local/blood supply , Neoplasm Recurrence, Local/metabolism , Aged , Aged, 80 and over , Antigens, Neoplasm/biosynthesis , Carbonic Anhydrase IX , Carbonic Anhydrases/biosynthesis , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Cell Hypoxia/physiology , Cell Line, Tumor , Endometrial Neoplasms/pathology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphatic Metastasis , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/metabolism
11.
Hum Reprod ; 22(1): 129-35, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16920727

ABSTRACT

BACKGROUND: Alterations in the progesterone receptor (PR) are considered a risk factor for the development of endometriosis. In this study, the frequencies of the PROGINS and +331G/A polymorphisms of the PR gene were determined in deep infiltrating endometriosis and correlated with the expression of the PR protein. METHODS AND RESULTS: The frequencies of the PR polymorphisms were determined in women with deep infiltrating endometriosis (n = 72), women with adenomyosis in the uterine wall (n = 40), gynaecological patients without symptomatic endometriosis (n = 102) and healthy females (n = 93). Detection of +331G/A and PROGINS polymorphisms was performed using PCR-restriction fragment length polymorphism (RFLP) analysis. Expression of PR-A and PR-B protein was assessed with immunohistochemistry. The allelic frequency of the polymorphic allele +331A was lower in women with endometriosis (P < 0.01) and adenomyosis (P < 0.02) compared with healthy females. The frequency of the PROGINS polymorphism did not differ between the groups. The mean staining index (SI) for PR-B in endometriotic epithelium was higher in the presence of the +331A polymorphic allele (n = 2) (P < 0.001) compared with +331G/G individuals (n = 61). The PROGINS polymorphism did not affect the SI for PR-A and PR-B. CONCLUSIONS: The presence of the PR gene polymorphic allele +331A is associated with a reduced risk of deep infiltrating endometriosis and adenomyosis compared with healthy population controls. The PROGINS polymorphism does not seem to modify the risk of deep infiltrating endometriosis.


Subject(s)
Endometriosis/genetics , Receptors, Progesterone/genetics , DNA Transposable Elements/genetics , Female , Gene Frequency , Humans , Polymorphism, Genetic , Risk Factors , Uterine Diseases/genetics
12.
Ann Oncol ; 18(3): 491-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17170014

ABSTRACT

BACKGROUND: Aberrations in mediators of Ras signaling may increase the risk of developing recurrent endometrial carcinoma. PATIENTS AND METHODS: Primary tumors of patients with (n = 44) and without (n = 44) recurrent stage I endometrioid endometrial carcinoma were compared regarding the presence of K-ras mutations (codons 12 and 13), B-raf mutations (V599), and RASSF1A gene promoter methylation. RESULTS: K-ras mutations were present in 18% of the patients independent of recurrent disease. No B-raf mutations were found. RASSF1A methylation was demonstrated in 85% of endometrial carcinomas, independent of recurrence. The presence of K-ras mutations and RASSF1A promoter methylation were not related, either directly or inversely. Analysis in premenopausal endometrial carcinomas demonstrated K-ras mutations in 40%, no B-raf mutations, and RASSF1A promoter methylation in 70% of the cases. RASSF1A methylation was also observed in samples of cyclic (n = 14), hyperplastic (n = 8), and atrophic (n = 13) endometrial tissues in 21%, 50% and 38%, respectively. CONCLUSIONS: RASSF1A methylation was observed in a high frequency in endometrioid endometrial carcinoma whereas K-ras and B-raf mutations were observed in a low frequency. No association was observed with the development of recurrent disease. High-frequency RASSF1A methylation in premenopausal carcinomas and an increased frequency in endometrial hyperplasia indicate that this may be an early event in endometrial carcinogenesis.


Subject(s)
Carcinoma, Endometrioid/genetics , DNA Methylation , Endometrial Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, ras , Neoplasm Recurrence, Local/genetics , Proto-Oncogene Proteins B-raf/genetics , Tumor Suppressor Proteins/genetics , Adult , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Case-Control Studies , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Endometrial Hyperplasia/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Middle Aged , Mutation , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Netherlands , Registries
13.
Gynecol Obstet Invest ; 62(4): 206-16, 2006.
Article in English | MEDLINE | ID: mdl-16785734

ABSTRACT

Infertility is an increasing problem all over the world, and it has been estimated that 10-15% of couples in fertile age have fertility problems. Likewise induced unsafe abortion is a serious threat to women's health. Despite advances made in assisted reproduction techniques, little progress has been made in increasing the success rate during fertility treatment. This document describes a wide range of projects carried out to increase the understanding in the field of embryo implantation research. The 'Fruitful' research network was created to encourage collaborations within the consortium and to describe our different research potentials to granting agencies or private sponsors.


Subject(s)
Embryo Implantation/physiology , Infertility, Female/physiopathology , Animals , Biomedical Research , Disease Models, Animal , Embryo Implantation/drug effects , Endometrium/physiology , Female , Humans , Pregnancy , Reproductive Techniques, Assisted , Trophoblasts/physiology
14.
J Steroid Biochem Mol Biol ; 100(4-5): 152-60, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16781141

ABSTRACT

In this study, we assessed the effects of tibolone and its metabolites on the production of a progesterone sensitive parameter, prolactin, in human endometrium stroma cells in vitro. In addition, the metabolism of the compounds by isolated stromal and epithelial cells was evaluated. The reference compounds, progesterone, Org 2058, and DHT all induced prolactin production. Oestradiol also slightly induced prolactin production and enhanced the response to Org 2058. Tibolone and Delta4-tibolone were similar with regard to potency to induce prolactin levels in the culture supernatant. Their potency was lower than that of Org 2058, similar to that of progesterone and higher than that of DHT. The efficacies of tibolone, Delta4-tibolone and Org 2058 were similar (approximately 200-fold induction). The estrogenic tibolone metabolites 3alpha- and 3beta-OH tibolone also significantly stimulated prolactin production. Their potency, however, was low since significance was reached only at the highest concentrations tested. The PR antagonist Org 31710 inhibited both tibolone- and Delta4-tibolone-induced prolactin production. The responses of tibolone and Delta4-tibolone were not affected by co-incubation with the androgen receptor antagonist OH-flutamide. The effect of tibolone, but not Delta4-tibolone, was antagonized approximately 50% in combination with the highest dose (1 microM) estrogen receptor antagonist, ICI 164384. The induction of prolactin by 3alpha- and 3beta-OH tibolone was antagonized most potently by Org 31710, but also by ICI 164384 and OH-flutamide. Tibolone is metabolized differently in epithelial and stromal cells of the human endometrium. The epithelial cells mostly produce the progestagenic/androgenic Delta4-tibolone. The stromal cells produce predominantly the 3beta-OH tibolone, and some Delta4-tibolone, but the net effect observed with regard to prolactin production is progestagenic. When the metabolites 3alpha-OH, 3beta-OH, and Delta4-tibolone were added to the cultures no conversions were observed. The HPLC analyses showed no evidence for the production of sulfated metabolites. In conclusion, the net effects on endometrial stromal cells are predominantly progestagenic. Tibolone is converted by epithelial cells into Delta4-tibolone which displays progestagenic and androgenic activities, whereas in stromal cells also the estrogenic metabolites 3alpha- and 3beta-OH tibolone are formed.


Subject(s)
Endometrium/cytology , Estrogen Receptor Modulators , Norpregnenes , Prolactin/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Cells, Cultured , Dihydrotestosterone/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estrogen Receptor Modulators/metabolism , Estrogen Receptor Modulators/pharmacology , Female , Humans , Norpregnenes/metabolism , Norpregnenes/pharmacology , Pregnenediones/chemistry , Pregnenediones/metabolism , Progesterone/chemistry , Progesterone/metabolism , Stromal Cells/cytology
15.
Mol Hum Reprod ; 12(6): 367-75, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16648151

ABSTRACT

Regeneration and growth of the human endometrium after shedding of the functional layer during menstruation depends on an adequate angiogenic response. We analysed the mRNA expression levels of all known vascular endothelial growth factor (VEGF) ligands and receptors in human endometrium collected in the menstrual and proliferative phases of the menstrual cycle. In addition, we evaluated the expression of VEGF-A, VEGF-R2 and NRP-1 at the protein level. Two periods of elevated mRNA expression of ligands and receptors were observed, separated by a distinct drop at cycle days (CDs) 9 and 10. Immunohistochemical staining showed that VEGF and VEGF-R2 were expressed in epithelial, stromal and endothelial cells. NRP-1 was mainly confined to stroma and blood vessels; only in late-proliferative endometrium, epithelial staining was also observed. Except for endothelial VEGF-R2 expression in CDs 6-8, there were no significant differences in the expression of VEGF, VEGF-R2 or NRP-1 in any of the cell compartments. In contrast, VEGF release by cultured human endometrium explants decreased during the proliferative phase. This output was significantly reduced in menstrual and early-proliferative endometrium by estradiol (E2) treatment. Western blot analysis indicated that part of the VEGF-A was trapped in the extracellular matrix (ECM). Changes in VEGF ligands and receptors were associated with elevated expression of the hypoxia markers HIF1alpha and CA-IX in the menstrual and early proliferative phases. HIF1alpha was also detected in late-proliferative phase endometrium. Our findings indicate that VEGF-A exerts its actions mostly during the first half of the proliferative phase. Furthermore, VEGF-A production appears to be triggered by hypoxia in the menstrual phase and subsequently suppressed by estrogen during the late proliferative phase.


Subject(s)
Endometrium/chemistry , Menstruation/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Blotting, Western , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Endometrium/cytology , Endometrium/metabolism , Female , Gene Expression/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Menstrual Cycle/genetics , Menstrual Cycle/metabolism , Menstruation/genetics , Middle Aged , Neuropilin-1/genetics , Neuropilin-1/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Angiogenesis ; 8(2): 147-56, 2005.
Article in English | MEDLINE | ID: mdl-16211360

ABSTRACT

Endometriosis, defined as the presence of endometrial tissue outside the uterus, is an estrogen-dependent disease which causes pelvic pain and subfertility in women of reproductive age. The condition has a dramatic impact on the professional, social and marital life of sufferers. Direct and indirect evidence suggests that angiogenesis is required for the development and persistence of endometriosis. In this review the state-of-the-art with regard to our understanding of the role of angiogenesis in the ectopic implantation and survival of menstrual endometrial tissue will be discussed.


Subject(s)
Blood Vessels/growth & development , Endometriosis/physiopathology , Endometrium/blood supply , Endometrium/pathology , Neovascularization, Pathologic/physiopathology , Blood Vessels/metabolism , Cell Movement/physiology , Female , Humans , Plasminogen Activators/metabolism , Plasminogen Inactivators/metabolism
17.
J Pathol ; 205(5): 597-605, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15726651

ABSTRACT

A case-control study was performed in order to determine whether expression of the progesterone receptor (PR) and/or aberrations of the PR gene contribute to the development of recurrent endometrial carcinoma. Primary tumours from 44 patients with recurrence of stage I endometrial carcinoma (patients) within 3 years after initial treatment were compared with tumours from 44 matched patients who were free of recurrence for a minimum of 3 years (controls). Paraffin wax-embedded primary tumours (n = 88) and recurrent tumours (n = 32) were analysed immunohistochemically for PR expression. A staining index (SI = 0-9) based on the staining intensity and the number of stained cells was calculated. DNA extracted from paraffin wax-embedded tissues was subjected to PCR-restriction fragment length polymorphism analysis (PCR-RFLP) for determination of the PROGINS DNA sequence alterations and the +331G/A-promoter polymorphism. Low PR expression (SI < 1.0) was observed in 7% of primary tumours derived from controls, 25% of primary tumours from patients with recurrence, and 38% of recurrent tumours. The expression of PR was significantly lower in primary tumours from patients with recurrence (SI = 4.0 +/- 0.5) than in the tumours in the control group (SI = 5.6 +/- 0.5) (T-test for paired analysis, p < 0.05). The PROGINS and +331G/A-promoter polymorphism were not related to age at diagnosis, tumour grade or myometrial invasion. The +331G/A-promoter polymorphism was present in 14% of primary tumours from patients without recurrence, compared with 17% of patients with recurrence. The PROGINS polymorphism was observed in 16% of primary tumours from patients without, and in 34% of patients with, recurrence (OR 2.6; 95% CI: 0.9-7.6). Most interestingly, patients who carried the PROGINS variant and in whom a PR-expressing tumour was diagnosed were at significantly enhanced risk of relapse (OR 4.7; 95% CI: 1.3-17.1). In conclusion, low PR expression tended to be associated with recurrent disease, and PR expression in tumours from patients carrying the PROGINS allele was predictive of the risk of recurrence.


Subject(s)
Biomarkers, Tumor/genetics , Chromosome Aberrations , Endometrial Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Receptors, Progesterone/genetics , Age Factors , Aged , Aged, 80 and over , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Case-Control Studies , DNA, Neoplasm/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/metabolism , Neoplasm Staging , Polymorphism, Genetic , Polymorphism, Restriction Fragment Length , Prognosis , Receptors, Progesterone/immunology , Receptors, Progesterone/metabolism
18.
Cell Mol Life Sci ; 62(2): 239-50, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15666095

ABSTRACT

To identify key regulatory mechanisms in the growth and development of the human endometrium, microarray analysis was performed on uncultured human endometrium collected during menstruation (M) and the late-proliferative (LATE-P)-phase of the menstrual cycle, as well as after 24 h incubation in the presence of oestradiol (17beta-E2). We demonstrate the expression of novel gene transcripts in the human endometrium. i.e. mucin-9, novel oestrogen-responsive gene transcripts, i.e. gelsolin and flotillin-1, and genes known to be expressed in human endometrium but not yet shown to be oestrogen responsive, i.e. connexin-37 and TFF1/pS2. Genes reported to be expressed during the implantation window and implicated in progesterone action, i.e. secretoglobin family 2A, member 2 (mammaglobin) and homeobox-containing proteins, were up-regulated in uncultured LATE-P-phase endometrium compared to M-phase endometrium. Some gene transcripts are regulated directly by 17beta-E2 alone, others are influenced by the in vivo environment as well. These observations emphasise that the regulation of endometrium maturation by oestrogen entails more then just stimulation of cell proliferation.


Subject(s)
Endometrium/metabolism , Estradiol/pharmacology , Gene Expression Regulation , Menstrual Cycle/physiology , Adult , Endometrium/cytology , Female , Gelsolin/genetics , Gene Expression Profiling , Glycoproteins/genetics , Humans , Membrane Proteins/genetics , Microarray Analysis , Middle Aged , Polymerase Chain Reaction
19.
J Steroid Biochem Mol Biol ; 92(3): 175-85, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15555911

ABSTRACT

The aim of this study was to evaluate the ex vivo oestrogen responsiveness of human proliferative phase endometrium using short-term explant cultures. The effects of oestrogen (17beta-E2) on proliferation and the expression of oestrogen-responsive genes known to be involved in regulating endometrial function were evaluated. Three distinct response patterns could be distinguished: (1) the menstrual (M) phase pattern (cycle days 2-5), which is characterised by a complete lack in the proliferative response to 17beta-E2, while an increased expression of AR (2.6-fold, P<0.01), PR (2.7-fold, P<0.01) and COX-2 (3.5-fold, P<0.01) at the mRNA level was observed and a similar upregulation was also found for AR, PR and COX-2 at the protein level; (2) the early proliferative (EP) phase pattern (cycle days 6-10) with 17beta-E2 enhanced proliferation in the stroma (1.7-fold, P<0.05), whereas the expression of AR, PR and COX-2 were not affected at the mRNA and protein levels and ER-alpha mRNA and protein levels were significantly reduced by 17beta-E2; (3) the late proliferative (LP) phase pattern (cycle days 11-14), which is characterised by a moderate stimulation of proliferation (1.4-fold, P<0.05) and PR mRNA expression (1.7-fold, P<0.01) by 17beta-E2. In conclusion, three distinct response patterns to 17beta-E2 could be identified with respect to proliferation and the expression of known oestrogen-responsive genes in human proliferative phase endometrium explant cultures.


Subject(s)
Endometrium/drug effects , Estrogens/pharmacology , Adult , Cell Division/drug effects , Cyclooxygenase 2 , Endometrium/cytology , Endometrium/growth & development , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Gene Expression/drug effects , Humans , Immunohistochemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Proteins , Middle Aged , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Tissue Culture Techniques
20.
Int J Gynecol Cancer ; 14(5): 947-56, 2004.
Article in English | MEDLINE | ID: mdl-15361208

ABSTRACT

Endometrial carcinoma, generally, has a good prognosis. However, in some patients, the tumor appears to behave very aggressively, a course that cannot be explained with histopathological characteristics. More insight into the molecular background can be valuable to clarify these differences in tumor behavior. The three components associated with the Wnt pathway--i.e., adenomatous polyposis coli (APC), beta-catenin, and E-cadherin--were evaluated in a case-control study of 28 patients with stage-I endometrial carcinomas to determine their involvement in the development of recurrent disease. Mutation analysis of the mutation cluster region of the APC gene, determination of gene promoter methylation status of the APC-1A and E-cadherin genes, and immunohistochemical analysis of APC, E-cadherin, and beta-catenin were performed using paraffin-embedded tumor tissue. Twenty-one APC gene mutations were detected in 12 of 28 (43%) patients. Only three mutations would result in a stopcodon in the APC gene. APC gene promoter methylation was assessed in 12 of 28 (43%) patients. APC immunostaining was absent in two of 24 (8.3%) patients. The occurrence of APC mutations, APC gene promoter methylation, and APC immunostaining were not predictive for recurrence. No E-cadherin expression was observed in four of 24 patients (17%). E-cadherin gene promoter methylation could not be detected in any of the patients. The absence of E-cadherin expression was predictive for distant metastases, but not for local recurrence. Nuclear localization of beta-catenin was present in nine of 24 (38%) patients and was not predictive for recurrent disease. Involvement of epigenetic and genetic aberrations in APC and beta-catenin genes seems to be of minor importance for the development of local recurrences and distant metastases. Although the number of patients is limited, E-cadherin expression appears to be predictive for the development of distant metastases in endometrial carcinoma.


Subject(s)
Cadherins/genetics , Carcinoma/genetics , Carcinoma/pathology , Cytoskeletal Proteins/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Genes, APC , Neoplasm Recurrence, Local/genetics , Trans-Activators/genetics , Adenomatous Polyposis Coli/genetics , Aged , Aged, 80 and over , Case-Control Studies , DNA Methylation , DNA Mutational Analysis , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Metastasis/genetics , Prognosis , Risk Factors , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...