Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Horm Cancer ; 2(3): 170-81, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21760855

ABSTRACT

Progesterone is a growth inhibitory hormone in the endometrium. While progestins can be used for the treatment of well-differentiated endometrial cancers, resistance to progestin therapy occurs for reasons that remain unclear. We have previously demonstrated that progesterone receptors (PR) A and B differentially regulate apoptosis in response to overexpression of the forkhead transcription factor, FOXO1. In this study, we further examined the PR-isoform-dependent cellular response to the AKT pathway. Treatment of PRA and PRB-expressing Ishikawa cells (PRA14, PRB23), with an AKT inhibitor API-59CJ-OMe (API-59) promoted apoptosis in the presence and absence of the ligand, R5020 preferentially in PRA14 cells. Upon PR knockdown using small interfering RNA, an increase in apoptosis was observed in PRB23 cells treated with API-59 with or without R5020 while there was no influence in PRA14 cells. Using an apoptosis-focused real-time PCR array, genes regulated by API-59 and R5020 were identified both common and unique to PRA14 and PRB23 cells. BIRC3 was identified as the only gene regulated by R5020 which occurred only in PRB cells. Knockdown of BIRC3 in PRB23 cells promoted a decrease in cell viability in response to API-59 + R5020. Furthermore, the important role of inhibitors of apoptosis (IAPs) in the PRB23 cells to promote cell survival was demonstrated using an antagonist to IAPs, a second mitochondria-derived activator of caspase (Smac also known as DIABLO) mimetic. Treatment of PRB23 cells with Smac mimetic increased apoptosis in response to API-59 + R5020. In summary, our findings indicate a mechanism by which PRB can promote cell survival in the setting of high AKT activity in endometrial cancer cells.


Subject(s)
Apoptosis/drug effects , Ellipticines/pharmacology , Endometrial Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/genetics , Inhibitor of Apoptosis Proteins/biosynthesis , Receptors, Progesterone/metabolism , Baculoviral IAP Repeat-Containing 3 Protein , Blotting, Western , Cell Line, Tumor , Cell Survival , Endometrial Neoplasms/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression , Humans , Inhibitor of Apoptosis Proteins/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Ubiquitin-Protein Ligases
2.
J Mol Histol ; 41(2-3): 111-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20383792

ABSTRACT

We have shown previously that high expression levels of microsomal epoxide hydrolase (mEH) correlate with a poor prognosis of breast cancer patients receiving tamoxifen, suggesting that enhanced mEH expression could lead to antiestrogen resistance (Fritz et al. in J Clin Oncol 19:3-9, 2001). Thus, the purpose of this study was to gain insights into the role of mEH in hormone-responsive tissues. We analyzed biopsy samples of the endometrium by immunohistochemical staining, pointing to a regulation of mEH during the menstrual cycle: during the first half mEH expression was low, increased during the second half and reached highest levels during pregnancy. Additionally, the progesterone receptor (PR) positive human endometrial cell lines IKPRAB-36 (estrogene receptor alpha [ERalpha] negative) and ECC1-PRAB72 (ERalpha positive) were chosen to further investigate the hormonal regulation of mEH expression. Western Blot and quantitative RT-PCR analysis revealed an increase of mEH expression after treatment with medroxy-progesterone 17-acetate (MPA) in the ERalpha containing ECC1-PRAB72 cells. In contrast our results suggest that MPA had no influence on the mEH protein level in the ERalpha- IKPRAB-36 cells. In conclusion, mEH expression is regulated by progesterone in the presence of both PRs and ERalpha.


Subject(s)
Endometrium/enzymology , Epoxide Hydrolases/biosynthesis , Gene Expression Regulation, Enzymologic , Menstrual Cycle/physiology , Progesterone/physiology , Blotting, Western , Cell Culture Techniques , Cell Line , Endometrium/cytology , Endometrium/drug effects , Endometrium/physiology , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Female , Humans , Immunohistochemistry , Medroxyprogesterone Acetate/pharmacology , Menstrual Cycle/drug effects , Receptors, Progesterone/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
3.
Clin Cancer Res ; 15(18): 5784-93, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19737954

ABSTRACT

PURPOSE: Wnt signaling regulates the fine balance between stemness and differentiation. Here, the role of Wnt signaling to maintain the balance between estrogen-induced proliferation and progesterone-induced differentiation during the menstrual cycle, as well as during the induction of hyperplasia and carcinogenesis of the endometrium, was investigated. EXPERIMENTAL DESIGN: Endometrial gene expression profiles from estradiol (E(2)) and E(2) + medroxyprogesterone acetate-treated postmenopausal patients were combined with profiles obtained during the menstrual cycle (PubMed; GEO DataSets). Ishikawa cells were transfected with progesterone receptors and Wnt inhibitors dickkopf homologue 1 (DKK1) and forkhead box O1 (FOXO1), measuring Wnt activation. Expression of DKK1 and FOXO1 was inhibited by use of sequence-specific short hairpins. Furthermore, patient samples (hormone-treated endometria, hyperplasia, and endometrial cancer) were stained for Wnt activation using nuclear beta-catenin and CD44. RESULTS: In vivo, targets and components of the Wnt signaling pathway (among them DKK1 and FOXO1) are regulated by E(2) and progesterone. In Wnt-activated Ishikawa cells, progesterone inhibits Wnt signaling by induction of DKK1 and FOXO1. Furthermore, using siRNA-mediated knockdown of both DKK1 and FOXO1, progesterone inhibition of Wnt signaling was partly circumvented. Subsequently, immunohistochemical analysis of the Wnt target gene CD44 showed that progesterone acted as an inhibitor of Wnt signaling in hyperplasia and in well-differentiated endometrial cancer. CONCLUSION: Progesterone induction of DKK1 and FOXO1 results in inhibition of Wnt signaling in the human endometrium. This Wnt inhibitory effect of progesterone is likely to play a rate-limiting role in the maintenance of endometrial homeostasis and, on its loss, in tumor onset and progression toward malignancy.


Subject(s)
Endometrial Neoplasms/metabolism , Endometrium/drug effects , Endometrium/metabolism , Progesterone/pharmacology , Signal Transduction/drug effects , Wnt Proteins/metabolism , beta Catenin/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Estrogens/metabolism , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Wnt Proteins/genetics , beta Catenin/genetics
4.
J Steroid Biochem Mol Biol ; 109(3-5): 219-23, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18434135

ABSTRACT

Tamoxifen is used as adjuvant treatment for postmenopausal breast cancer patients. The mechanism of action of tamoxifen in breast cancer patients is that tamoxifen inhibits growth of cancer cells by competitive antagonism for estrogens at the estrogen receptor (ER). In the endometrium, tamoxifen has an effect that varies with the ambient concentration of estrogen: in premenopausal women (high estrogen levels), tamoxifen displays an estrogen-antagonistic effect, while in postmenopausal women (low estrogen levels), tamoxifen displays an estrogen-agonistic mode of action. Here, using microarray technology we have compared estrogen signaling with tamoxifen signaling in the human endometrium. It was observed that on the one hand tamoxifen-treatment results in modulation of expression of specific genes (370 genes) and on the other hand tamoxifen-treatment results in modulation of a set of genes which are also regulated by estrogen treatment (142 genes). Upon focusing on regulation of proliferation, we found that tamoxifen-induced endometrial proliferation is largely accomplished by using the same set of genes as are regulated by estradiol. So, as far as regulation of proliferation goes, tamoxifen seems to act as estrogen agonist. Furthermore, tamoxifen-specific gene regulation may explain why tamoxifen-induced endometrial tumors behave more aggressively than sporadic endometrial tumors.


Subject(s)
Endometrium/drug effects , Endometrium/metabolism , Estrogens/metabolism , Signal Transduction/drug effects , Tamoxifen/pharmacology , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Polyps/pathology
5.
Hum Reprod ; 23(2): 298-305, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077316

ABSTRACT

BACKGROUND: Combined hormone treatments in post-menopausal women have different clinical responses on uterus and vagina; therefore, we investigated differences in steroid signalling between various hormone therapies in these tissues. METHODS: A total of 30 post-menopausal women scheduled for hysterectomy were distributed into four subgroups: control-group (n = 9), Tibolone-group (n = 8); estradiol (E(2))-group (n = 7); E(2) + medroxyprogesterone acetate (MPA)-group (n = 6). Medication was administered orally every day for 21 days prior to removal of uterus and upper part of the vagina. Tissue RNA was isolated, and gene expression profiles were generated using GeneChip technology and analysed by cluster analysis and significance analysis of microarrays. Apoptosis and cell proliferation assays, as well as immunohistochemistry for hormone receptors were performed. RESULTS: 21-days of treatment with E(2), E(2) + MPA or tibolone imposes clear differential gene expression profiles on endometrium and myometrium. Treatment with E(2) only results in the most pronounced effect on gene expression (up to 1493 genes differentially expressed), proliferation and apoptosis. Tibolone, potentially metabolized to both estrogenic and progestagenic metabolites, shows some resemblance to E(2) signalling in the endometrium and, in contrast, shows significant resemblance to E(2) + MPA signalling in the myometrium. In the vagina the situation is entirely different; all three hormonal treatments result in regulation of a small number (4-73) of genes, in comparison to signalling in endometrium and myometrium. CONCLUSION: Endometrium and myometrium differentially respond to the hormone therapies and use completely different sets of genes to regulate similar biological processes, while in this experiment the upper part of the vagina is hardly hormone responsive.


Subject(s)
Endometrium/metabolism , Estrogen Replacement Therapy , Myometrium/metabolism , Signal Transduction/drug effects , Vagina/metabolism , Cluster Analysis , Drug Therapy, Combination , Estradiol/therapeutic use , Female , Gene Expression/drug effects , Gene Expression Profiling , Humans , Medroxyprogesterone Acetate/therapeutic use , Norpregnenes/therapeutic use
6.
Reprod Sci ; 14(7): 646-54, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18000226

ABSTRACT

For the endometrium, estradiol and tamoxifen induce proliferation, and consequently, tamoxifen treatment of breast cancer results in a 2-fold to 7-fold increased risk for endometrial cancer. Here, the role of activation of growth factor receptor signaling in mediating the effects of estrogen and tamoxifen is determined. Microarray analysis of ECC-1 cells treated with estradiol or tamoxifen indicate that rapid responses to treatment (1 hour) are very distinct from long-term responses (>24 hours). Furthermore, estradiol and tamoxifen are observed to induce AKT activation. Comparing long-term estrogen- and tamoxifen-regulated genes with genes regulated by insulin-like growth factor 1 and amphiregulin reveals that the late effects of estrogen and tamoxifen signaling may partly be mediated via activation of growth factor receptor signaling pathways. It is hypothesized that both early and late effects of estrogen and tamoxifen signaling in the endometrium are partly mediated via the activation of growth factor receptor signaling, putatively at the level of AKT activation.


Subject(s)
Endometrium/metabolism , Estradiol/pharmacology , Estrogens/physiology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Growth Factor/metabolism , Tamoxifen/pharmacology , Cell Line, Tumor , Endometrium/drug effects , ErbB Receptors/metabolism , Estrogens/genetics , Estrogens/pharmacology , Female , Gene Expression Profiling , Humans , Insulin-Like Growth Factor I/metabolism , Multigene Family , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-akt/genetics , Receptors, Growth Factor/genetics , Signal Transduction
7.
Reprod Sci ; 14(2): 160-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17636227

ABSTRACT

Tibolone has estrogenic effects on the vagina but not on the uterus. To explain this, levels of tibolone and estradiol and their metabolites were determined in serum, myometrium, and vagina. Thirty-four postmenopausal women with uterine prolapse received either no treatment, tibolone, E(2) or E(2) + medroxyprogesterone acetate (MPA) for 21 days, or a single dose of tibolone. Twenty +/- 6 hours after administration, >98% of the 3-hydroxytibolone metabolites in serum and tissues were disulfated. Of the unconjugated metabolites, the estrogenic 3alpha-hydroxytibolone predominated in serum, whereas the progestagenic/ androgenic Delta(4)-tibolone predominated in myometrium and vagina. Levels of disulfated metabolites in serum and tissues were higher (3- to 5-fold) after multiple dosing than after a single dose. Tissue:serum ratios were <1, except for Delta(4)-tibolone. In all groups, E(2) tissue levels were higher than serum levels; the percentage of serum E(1)S was >90%. Tibolone did not affect endogenous E(1), E(2), or E(1)S levels in serum, but in myometrium and vagina, E(1) levels were significantly higher and E(1)S levels tended to be lower than in controls. Serum and tissue levels of endogenous and exogenous E(1), E(2), and E(1)S were markedly increased 20 hours after E(2) or E(2) + MPA; the percentage of E(1)S and tissue:serum ratios were not affected. MPA had no effect on the degree of sulfation of E(1). Compared with serum, tissue levels of E(2) were high in all groups; absolute E(2) levels in control and tibolone groups were much lower than in the E(2) groups. Tibolone metabolite patterns are different in serum, myometrium, and vagina.


Subject(s)
Estradiol/metabolism , Estrone/analogs & derivatives , Medroxyprogesterone Acetate/metabolism , Myometrium/metabolism , Norpregnenes/metabolism , Selective Estrogen Receptor Modulators/metabolism , Vagina/metabolism , Aged , Estradiol/administration & dosage , Estradiol/blood , Estrone/blood , Estrone/metabolism , Female , Humans , Medroxyprogesterone Acetate/administration & dosage , Medroxyprogesterone Acetate/blood , Middle Aged , Myometrium/drug effects , Norpregnenes/administration & dosage , Norpregnenes/blood , Postmenopause/blood , Postmenopause/metabolism , Selective Estrogen Receptor Modulators/administration & dosage , Selective Estrogen Receptor Modulators/blood , Tissue Distribution , Uterine Prolapse/blood , Uterine Prolapse/metabolism , Uterine Prolapse/surgery , Vagina/drug effects
8.
Int J Cancer ; 121(4): 759-66, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17471573

ABSTRACT

UNLABELLED: Vulvar intraepithelial neoplasia (VIN) is a premalignant disorder caused by human papillomaviruses. Basic knowledge about the molecular pathogenesis of VIN is sparse. Therefore, we have analyzed the gene expression profile of 9 VIN samples in comparison to 10 control samples by using genome wide Affymetrix Human U133A plus2 GeneChips. Results were validated by quantitative real-time RT-PCR analysis and immunostaining of a few representative genes (TACSTD1, CCNE2, AR and ESR1). Significance analysis of microarrays (SAM) showed that 1,497 genes were differentially expressed in VIN compared to controls. By analyzing the biological processes affected by the observed differences, we found that VIN appears to be a highly proliferative disease; many cyclins (CCNA, CCNB and CCNE) and almost all prereplication complex proteins are upregulated. Thereby, VIN does not seem to depend for its proliferation on paracrine or endocrine signals. Many receptors (for example ESR1 and AR) and ligands are downregulated. Furthermore, although VIN is not an invasive disease, the inhibition of expression of a marked number of cell-cell adhesion molecules seems to indicate development towards invasion. Upon reviewing apoptosis and angiogenesis, it was observed that these processes have not become significantly disregulated in VIN. IN CONCLUSION: although VIN is still a premalignant disease, it already displays several hallmarks of cancer.


Subject(s)
Alphapapillomavirus , Carcinoma in Situ/virology , Precancerous Conditions/virology , Signal Transduction , Vulvar Neoplasms/virology , Adult , Carcinoma in Situ/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Female , Gene Expression Profiling , Humans , Middle Aged , Precancerous Conditions/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Vulvar Neoplasms/genetics
9.
Fertil Steril ; 86(2): 352-61, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16828477

ABSTRACT

OBJECTIVE: To evaluate histological and immunohistochemical parameters of short-term (21 days) tibolone, estrogen-only, and estrogen+progestagen treatment in the human postmenopausal endometrium. DESIGN: An observational, open, nonrandomized, controlled study. SETTING: Three collaborating centers: Amphia Hospital in Breda, Albert Schweitzer Hospital in Dordrecht, Erasmus Medical Center in Rotterdam, the Netherlands. PATIENT(S): Thirty healthy, postmenopausal women. INTERVENTION(S): Control group (n = 9), no hormonal treatment; tibolone group (n = 8), patients were treated with 2.5 mg of tibolone (administered orally) every day, starting 21 days before surgery; estrogen group (n = 7), patients were treated with 2 mg of E(2) (Zumenon, administered orally; Zambon, Amerfoort; The Netherlands) every day, starting 21 days before surgery; estrogen+progestagen group (n = 6), patients were treated with 2 mg of E(2) (Zumenon, administered orally) and 5 mg of medroxyprogesterone acetate (administered orally) every day, starting 21 days before surgery. MAIN OUTCOME MEASURE(S): Uterine tissues were collected, and two pathologists independently assessed histology. Immunohistochemical parameters measured were estrogen receptor alpha, progesterone receptor A/B, Hoxa10, Ki67, and Bcl-2. RESULT(S): On the basis of a number of histological and immunohistochemical parameters measured after 21 days of treatment, it was observed that tibolone displays clearly less stimulation (proliferation) of the human postmenopausal endometrium than estrogen at the beginning of a treatment, but the stimulation is higher than with estrogen+progestagen. CONCLUSION(S): Short-term (21 days) tibolone treatment results in a small stimulation of proliferation of the endometrium, and because long-term treatment with tibolone has been demonstrated to lead to an atrophic endometrium, it may be concluded that the stimulatory effect, as observed in this study, is transient in nature. It is hypothesized that tibolone first displays a more estrogenic mode of action, which over time, is counterbalanced by the induction of its progestagenic properties.


Subject(s)
Cell Proliferation/drug effects , Endometrium/metabolism , Endometrium/pathology , Estrogens/pharmacology , Norpregnenes/pharmacology , Postmenopause/metabolism , Progestins/pharmacology , Aged , Drug Synergism , Female , Humans , Hysterectomy , Immunohistochemistry , Middle Aged , Mitosis/drug effects , Uterine Prolapse/metabolism , Uterine Prolapse/pathology , Uterine Prolapse/surgery
10.
J Soc Gynecol Investig ; 12(7): e55-67, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16202921

ABSTRACT

OBJECTIVES: In different tissues, estrogens, selective estrogen receptor modulators (SERMs), and anti-estrogens exert different biologic activities. For the endometrium, estradiol and tamoxifen induce proliferation, and because of this, tamoxifen treatment of breast cancer patients results in a two- to sevenfold increased risk for development of endometrial cancer. Use of raloxifene, or the anti-estrogen ICI182780, does not result in such an increased risk. The objective of the current study was to generate and analyze gene expression profiles that reflect the transcriptional response of the human endometrium to estradiol, SERMs like tamoxifen and raloxifene, and anti-estrogens like ICI182780. METHODS: Transient transfections were performed to analyze the transcriptional response of ECC-1 cells to estradiol, tamoxifen, raloxifene, and ICI182780. Subsequently, to reveal the molecular mechanism of action, gene expression profiles were generated and some of the observed regulated genes were confirmed by Northern blotting. Biostatistical methods were employed to analyze the expression profile results further, and amphiregulin effects on ECC-1 cell signaling were investigated using Northern and Western blotting, and 3H-thymidine incorporation. RESULTS: Analysis of the profiles revealed that estradiol, tamoxifen, raloxifene, and ICI182780 influence the same biologic processes, but they do so via regulation of different sets of genes. Upon construction of a genetic network it was observed that the largest possible network centered on epidermal growth factor (EGF) receptor signaling. Furthermore, the EGF receptor ligand amphiregulin was differentially regulated by all four ligands. Next it was shown that amphiregulin indeed could stimulate EGF receptor signaling in ECC-1 cells. Based on these results, it was hypothesized that EGF receptor signaling could differentially be affected by estrogen, tamoxifen, raloxifene, and ICI182780 because these four compounds differentially regulate the EGF receptor ligand amphiregulin. CONCLUSIONS: Regulation of amphiregulin coincides with the described in vivo effect of the four ligands on the endometrium. Therefore, it is possible that modulation of EGF receptor signaling is a significant player in estrogen-agonistic growth of the endometrium and needs to be investigated further.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Endometrial Neoplasms/pathology , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Glycoproteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Amphiregulin , EGF Family of Proteins , Estradiol/pharmacology , Estradiol/physiology , Female , Fulvestrant , Gene Expression Profiling , Humans , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...