82 results on '"Blok LJ"'
Search Results
2. The psychological impact of gestational trophoblastic disease: a prospective observational multicentre cohort study.
- Author
-
Blok, LJ, Frijstein, MM, Eysbouts, YK, Custers, JAE, Sweep, FCGJ, Lok, CAR, and Ottevanger, PB
- Subjects
- *
PSYCHOLOGICAL factors , *PSYCHOLOGICAL distress , *IMPACT of Event Scale , *MOLAR pregnancy , *GESTATIONAL trophoblastic disease , *MISCARRIAGE , *COHORT analysis - Abstract
Objective: To evaluate the short‐term psychological consequences of gestational trophoblastic disease (GTD). Design: A prospective observational multicentre cohort study. Setting: Nationwide in the Netherlands. Population: GTD patients. Methods: Online questionnaires directly after diagnosis. Main outcome measures: Hospital Anxiety and Depression Scale (HADS), Distress Thermometer (DT), Impact of Event Scale (IES) and Reproductive Concerns Scale (RCS). Results: Sixty GTD patients were included between 2017 and 2020. Anxious feelings (47%) were more commonly expressed than depressive feelings (27%). Patients experienced moderate to severe adaptation problems in 88%. Patients who already had children were less concerned about their reproductivity than were patients without children (mean score 10.4 versus 15.0, P = 0.031), and patients with children experienced lower distress levels (IES mean score 25.7 versus 34.7, P = 0.020). In addition, patients with previous pregnancy loss scored lower for distress compared with patients without pregnancy loss (IES mean score 21.1 versus 34.2, P = 0.002). Discussion: We recommend that physicians monitor physical complaints and the course of psychological wellbeing over time in order to provide personalised supportive care in time for patients who have high levels of distress at baseline. Conclusions: GTD patients experience increased levels of distress, anxiety and depression, suggesting the diagnosis has a substantial effect on the psychological wellbeing of patients. The impact of GTD diagnosis on intrusion and avoidance seems to be ameliorated in patients who have children or who have experienced previous pregnancy loss. Patients with gestational trophoblastic disease (GTD) experience short‐term psychological consequences such as distress, anxiety and depression, suggesting that the diagnosis GTD has a substantial effect on the psychological wellbeing of patients. Various patient characteristics affect the impact of GTD diagnosis. Patients with gestational trophoblastic disease (GTD) experience short‐term psychological consequences such as distress, anxiety and depression, suggesting that the diagnosis GTD has a substantial effect on the psychological wellbeing of patients. Various patient characteristics affect the impact of GTD diagnosis. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
3. Authors' response re: The psychological impact of gestational trophoblastic disease: a prospective observational multicentre cohort study.
- Author
-
Blok LJ, Frijstein MM, Eysbouts YK, Custers JA, Sweep FC, Lok CA, and Ottevanger PB
- Subjects
- Cohort Studies, Female, Humans, Pregnancy, Prospective Studies, Gestational Trophoblastic Disease, Hydatidiform Mole
- Published
- 2022
- Full Text
- View/download PDF
4. Progesterone-induced miR-133a inhibits the proliferation of endometrial epithelial cells.
- Author
-
Pan JL, Yuan DZ, Zhao YB, Nie L, Lei Y, Liu M, Long Y, Zhang JH, Blok LJ, Burger CW, and Yue LM
- Subjects
- Animals, Blotting, Western, Cell Proliferation drug effects, Cell Proliferation physiology, Epithelial Cells metabolism, Female, Flow Cytometry, Immunohistochemistry, Mice, Real-Time Polymerase Chain Reaction, Cyclin D2 biosynthesis, Endometrium metabolism, Gene Expression Regulation drug effects, MicroRNAs metabolism, Progesterone pharmacology
- Abstract
Aim: This study aimed to understand the role of miR-133a in progesterone actions, explore the regulative mechanism of the progesterone receptor, and investigate the effects of miR-133a on the progesterone-inhibited proliferation of mouse endometrial epithelial cells., Methods: The expression of miR-133a induced by progesterone was detected by quantitative real-time PCR both in vivo and in vitro. Ishikawa subcell lines stably transfected with progesterone receptor subtypes were used to determine the receptor mechanism of progesterone inducing miR-133a. Specific miR-133a mimics or inhibitors were transfected into mouse uteri and primary cultured endometrial epithelial cells to overexpress or downregulate the miR-133a. The roles of miR-133a in the cell cycle and proliferation of endometrial epithelial cells were analysed by flow cytometry and Edu incorporation analysis. The protein levels of cyclinD2 in uterine tissue sections and primary cultured endometrial epithelial cells were determined by immunohistochemistry and Western blot analysis., Results: Progesterone could induce miR-133a expression in a PRB-dependent manner in endometrial epithelial cells. miR-133a inhibited endometrial epithelial cell proliferation by arresting cell cycle at the G
1 -S transition. Moreover, miR-133a acted as an inhibitor in downregulating cyclinD2 in endometrial epithelial cells., Conclusion: We showed for the first time that progesterone-induced miR-133a inhibited the proliferation of endometrial epithelial cells by downregulating cyclinD2. Our research indicated an important mechanism for progesterone inhibiting the proliferation of endometrial epithelial cells by inducing special miRNAs to inhibit positive regulatory proteins in the cell cycle., (© 2016 Scandinavian Physiological Society. Published by John Wiley & Sons Ltd.)- Published
- 2017
- Full Text
- View/download PDF
5. IL6/JAK1/STAT3 Signaling Blockade in Endometrial Cancer Affects the ALDHhi/CD126+ Stem-like Component and Reduces Tumor Burden.
- Author
-
van der Zee M, Sacchetti A, Cansoy M, Joosten R, Teeuwssen M, Heijmans-Antonissen C, Ewing-Graham PC, Burger CW, Blok LJ, and Fodde R
- Subjects
- Aldehyde Dehydrogenase genetics, Animals, Cell Line, Tumor, Cell Proliferation drug effects, Cisplatin administration & dosage, Endometrial Neoplasms drug therapy, Endometrial Neoplasms pathology, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Interleukin-6 genetics, Janus Kinase 1 genetics, Mice, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Receptors, Interleukin-6 antagonists & inhibitors, STAT3 Transcription Factor genetics, Signal Transduction drug effects, Tumor Burden genetics, Aldehyde Dehydrogenase biosynthesis, Endometrial Neoplasms genetics, Interleukin-6 biosynthesis, Janus Kinase 1 biosynthesis, STAT3 Transcription Factor biosynthesis
- Abstract
Cancer stem-like cells (CSC) may be critical to maintain the malignant behavior of solid and hematopoietic cancers. Recently, patients with endometrial cancer whose tumors expressed high levels of aldehyde dehydrogenase (ALDH), a detoxifying enzyme characteristic of many progenitor and stem cells, exhibited a relative reduction in survival compared with patients with low levels of ALDH. Given evidence of its role as a CSC marker, we hypothesized that high level of ALDH activity (ALDH(hi)) in a tumor might positively correlate with the presence of stem- and progenitor-like tumor cells in this disease setting. In support of this hypothesis, ALDH could be used to enrich for CSC in endometrial cancer cell lines and primary tumors, as illustrated by the increased tumor-initiating capacity of ALDH(hi) cells in immunodeficient mice. ALDH(hi) cells also exhibited greater clonogenic and organoid-forming capacity compared with ALDH(lo) cells. Notably, the number of ALDH(hi) cells in tumor cell lines and primary tumors inversely correlated with differentiation grade. Expression analysis revealed upregulation of IL6 receptor subunits and signal transducers CD126 and GP130 in ALDH(hi) endometrial cancer cells. Accordingly, targeted inhibition of the IL6 receptor and its downstream effectors JAK1 and STAT3 dramatically reduced tumor cell growth. Overall, our results provide a preclinical rationale to target IL6 or its effector functions as a novel therapeutic option in endometrial cancer., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
6. Progesterone receptor membrane component 1 deficiency attenuates growth while promoting chemosensitivity of human endometrial xenograft tumors.
- Author
-
Friel AM, Zhang L, Pru CA, Clark NC, McCallum ML, Blok LJ, Shioda T, Peluso JJ, Rueda BR, and Pru JK
- Subjects
- Animals, Blotting, Western, Endometrial Neoplasms pathology, Female, Humans, Immunoenzyme Techniques, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Mice, Mice, Inbred NOD, Mice, SCID, Mitosis, RNA, Messenger genetics, RNA, Small Interfering genetics, Real-Time Polymerase Chain Reaction, Receptors, Progesterone antagonists & inhibitors, Receptors, Progesterone genetics, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Apoptosis, Cell Proliferation, Drug Resistance, Neoplasm, Endometrial Neoplasms metabolism, Endometrial Neoplasms prevention & control, Membrane Proteins metabolism, Receptors, Progesterone metabolism
- Abstract
Endometrial cancer is the leading gynecologic cancer in women in the United States with 52,630 women predicted to be diagnosed with the disease in 2014. The objective of this study was to determine if progesterone (P4) receptor membrane component 1 (PGRMC1) influenced endometrial cancer cell viability in response to chemotherapy in vitro and in vivo. A lentiviral-based shRNA knockdown approach was used to generate stable PGRMC1-intact and PGRMC1-deplete Ishikawa endometrial cancer cell lines that also lacked expression of the classical progesterone receptor (PGR). Progesterone treatment inhibited mitosis of PGRMC1-intact, but not PGRMC1-deplete cells, suggesting that PGRMC1 mediates the anti-mitotic actions of P4. To test the hypothesis that PGRMC1 attenuates chemotherapy-induced apoptosis, PGRMC1-intact and PGRMC1-deplete cells were treated in vitro with vehicle, P4 (1 µM), doxorubicin (Dox, 2 µg/ml), or P4 + Dox for 48 h. Doxorubicin treatment of PGRMC1-intact cells resulted in a significant increase in cell death; however, co-treatment with P4 significantly attenuated Dox-induced cell death. This response to P4 was lost in PGRMC1-deplete cells. To extend these observations in vivo, a xenograft model was employed where PGRMC1-intact and PGRMC1-deplete endometrial tumors were generated following subcutaneous and intraperitoneal inoculation of immunocompromised NOD/SCID and nude mice, respectively. Tumors derived from PGRMC1-deplete cells grew slower than tumors from PGRMC1-intact cells. Mice harboring endometrial tumors were then given three treatments of vehicle (1:1 cremophor EL: ethanol + 0.9% saline) or chemotherapy [Paclitaxel (15 mg/kg, i.p.) followed after an interval of 30 minutes by CARBOplatin (50 mg/kg)] at five day intervals. In response to chemotherapy, tumor volume decreased approximately four-fold more in PGRMC1-deplete tumors when compared with PGRMC1-intact control tumors, suggesting that PGRMC1 promotes tumor cell viability during chemotherapeutic stress. In sum, these in vitro and in vivo findings demonstrate that PGRMC1 plays a prominent role in the growth and chemoresistance of human endometrial tumors., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
7. Progesterone-induced cyclin G1 inhibits the proliferation of endometrial epithelial cell and its possible molecular mechanism.
- Author
-
Yuan DZ, Ding XL, Yu HL, Cheng Z, Tang XR, He YP, Zhang JH, Blok LJ, Hanifi-Moghaddam P, Burger CW, and Yue LM
- Subjects
- Animals, Cell Proliferation drug effects, Epithelial Cells cytology, Epithelial Cells drug effects, Female, Humans, Mice, Okadaic Acid pharmacology, Protein Binding drug effects, Protein Phosphatase 2 metabolism, RNA, Small Interfering metabolism, Cyclin G1 metabolism, Endometrium cytology, Epithelial Cells metabolism, Progesterone pharmacology
- Abstract
Under normal conditions, progesterone inhi-bits the estrogen-induced proliferation of endometrial epithelium. Our previous studies have shown that cyclin G1 was progesterone-dependent in mouse endometrial epithelium at peri-implantation, and exogenous cyclin G1 suppressed the proliferation of endometrial cancer cells. The objectives of this study are to determine whether cyclin G1, as a negative regulator of the cell cycle, is involved in the antiproliferative action of progesterone on endometrial epithelial cells, and to explore the possible molecular mechanism of cyclin G1 inhibition. The siRNA-mediated elimination of cyclin G1 attenuated the antiproliferative action of progesterone on endometrial epithelial cells. Immunoprecipitation showed that progesterone-induced cyclin G1 could interact with PP2A to mediate its phosphatase activity. The block of PP2A activity also attenuated the antiproliferative action of progesterone on endometrial epithelial cells and increased the phosphorylated Rb. In conclusion, progesterone-induced cyclin G1 mediates the inhibitory effect of progesterone on endometrial epithelial cell proliferation possibly through the recruitment of PP2A to dephosphorylate Rb., (© Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2014
- Full Text
- View/download PDF
8. A mouse model for endometrioid ovarian cancer arising from the distal oviduct.
- Author
-
van der Horst PH, van der Zee M, Heijmans-Antonissen C, Jia Y, DeMayo FJ, Lydon JP, van Deurzen CH, Ewing PC, Burger CW, and Blok LJ
- Subjects
- Adenomatous Polyposis Coli genetics, Animals, Carcinoma, Endometrioid genetics, Carcinoma, Ovarian Epithelial, Endometrial Hyperplasia pathology, Endometrium pathology, Epithelial Cells pathology, Female, Mice, Inbred C57BL, Mice, Knockout, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Neoplasms, Glandular and Epithelial genetics, Ovarian Neoplasms genetics, Wnt Proteins metabolism, beta Catenin metabolism, Carcinoma, Endometrioid pathology, Disease Models, Animal, Fallopian Tubes pathology, Mice, Neoplasms, Glandular and Epithelial pathology, Ovarian Neoplasms pathology, Wnt Signaling Pathway genetics
- Abstract
Ovarian cancer is the deadliest gynecological malignancy in Western countries. Early detection, however, is hampered by the fact that the origin of ovarian cancer remains unclear. Knowing that in a high percentage of endometrioid ovarian cancers Wnt/β-catenin signaling is activated, and in view of the hypothesis that ovarian cancer may originate from the distal oviduct, we studied mice in which Wnt/β-catenin signaling was activated in Müllerian duct-derived tissues. Conditional adenomatous polyposis coli (Apc) knockout mice were used to study the activation of Wnt/β-catenin signaling in Müllerian duct-derived organs. These Pgr(Cre/+);Apc(ex15lox/lox) mice (n = 44) were sacrificed at 10, 20, 40 and 80 weeks and uterus, oviduct, ovaries and surrounding fat tissues were assessed using immunohistochemistry. Using nuclear β-catenin staining, Wnt/β-catenin signaling activation was confirmed in the entire epithelium of the adult Müllerian duct (fimbriae, oviduct and endometrium), but was absent in ovarian surface epithelium cells (OSEs). Besides endometrial hyperplasia, in 87.2% of mice intraepithelial lesions of the distal oviduct were found, whereas OSEs remained unaffected. In addition, 62.5% of mice developed tumors in the distal and fimbrial part of the oviduct. In the ovaries, mainly at young age, in 16.3% of mice, simple epithelial cysts were noted, which developed further into endometrioid ovarian tumors, resembling human endometrioid ovarian cancer (27.9% of mice). Next to this, locoregional growth in the utero-ovarian ligament was also shown. Here, for the first time, mutations (activation of Wnt/β-catenin) in the distal oviduct result in precursor lesions that develop into ovarian tumors, resembling human endometrioid ovarian cancer., (© 2014 UICC.)
- Published
- 2014
- Full Text
- View/download PDF
9. Alterations in Wnt-β-catenin and Pten signalling play distinct roles in endometrial cancer initiation and progression.
- Author
-
van der Zee M, Jia Y, Wang Y, Heijmans-Antonissen C, Ewing PC, Franken P, DeMayo FJ, Lydon JP, Burger CW, Fodde R, and Blok LJ
- Subjects
- Animals, Carcinoma, Squamous Cell pathology, Disease Progression, Endometrial Neoplasms pathology, Female, Gene Deletion, Gene Silencing, Genes, APC physiology, Loss of Heterozygosity genetics, Male, Mice, Mice, Knockout, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, Uterus abnormalities, Uterus metabolism, Carcinoma, Squamous Cell metabolism, Endometrial Neoplasms metabolism, PTEN Phosphohydrolase metabolism, Wnt Signaling Pathway physiology, beta Catenin metabolism
- Abstract
Endometrioid endometrial cancer arises through a gradual series of histological changes, each accompanied by specific alterations in gene expression and activity. Activation of the Wnt-β-catenin pathway and loss of PTEN activity are frequently observed in endometrial cancers. However, the specific roles played by alterations in these pathways in the initiation and progression of endometrial cancer are currently unclear. Here, we investigated the effects of loss of Pten and Apc gene function in the mouse endometrium by employing tissue-specific and inducible mutant alleles, followed by immunohistochemical (IHC) and loss of heterozygosity (LOH) analysis of their corresponding cancerous lesions. Loss of the Apc function in the endometrium leads to cytoplasmic and nuclear β-catenin accumulation in association with uterine hyperplasia and squamous cell metaplasia, but without malignant transformation. Loss of Pten function also resulted in squamous metaplasia but, in contrast to loss of Apc function, it initiates endometrial cancer. On the other hand, loss of Apc function in the endometrium accelerates Pten-driven endometrial tumourigenesis. Analysis of compound heterozygous mice confirmed that somatic loss of the wild-type Pten allele represents the rate-limiting initiation step in endometrial cancer. Simultaneous loss of Pten and Apc resulted in endometrial cancer characterized by earlier onset and a more aggressive malignant behaviour. These observations are indicative of the synergistic action between the Wnt-β-catenin and Pten signalling pathways in endometrial cancer onset and progression., (Copyright © 2013 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
10. Interaction between sex hormones and WNT/β-catenin signal transduction in endometrial physiology and disease.
- Author
-
van der Horst PH, Wang Y, van der Zee M, Burger CW, and Blok LJ
- Subjects
- Animals, Embryo Implantation physiology, Female, Humans, Placentation, Pregnancy, Endometrium metabolism, Endometrium physiopathology, Gonadal Steroid Hormones metabolism, Uterine Diseases metabolism, Uterine Diseases physiopathology, Wnt Signaling Pathway
- Abstract
Wnt/β-catenin signalling plays a rate-limiting role in early development of many different organs in a broad spectrum of organisms. In the developing Müllerian duct, Wnt/β-catenin signalling is important for initiation, outgrowth, patterning and differentiation into vagina, cervix, uterus and oviducts. In adult life, sex hormones modulate Wnt/β-catenin signalling in the endometrium to maintain the monthly balance between estrogen-induced proliferation and progesterone-induced differentiation, and enhanced Wnt/β-catenin signalling seems to be involved in endometrial carcinogenesis. However, early in pregnancy enhanced Wnt/β-catenin signalling is prerequisite for proper implantation and invasion of trophoblast cells into endometrium and myometrium thus helping to form a placenta. Overall, it seems that tight control of Wnt/β-catenin signalling in time and space is important for initiation, development and normal function of the female reproductive tract. However, if Wnt/β-catenin signalling is not kept in check, it easily seems to initiate or contribute to development of a number of uterine disorders., (Copyright © 2011 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
11. Different DNA damage and cell cycle checkpoint control in low- and high-risk human papillomavirus infections of the vulva.
- Author
-
Santegoets LA, van Baars R, Terlou A, Heijmans-Antonissen C, Swagemakers SM, van der Spek PJ, Ewing PC, van Beurden M, van der Meijden WI, Helmerhorst TJ, and Blok LJ
- Subjects
- BRCA1 Protein biosynthesis, Biomarkers, Tumor, Condylomata Acuminata genetics, Condylomata Acuminata metabolism, Condylomata Acuminata pathology, Condylomata Acuminata virology, Cyclin-Dependent Kinase Inhibitor p16 metabolism, DNA, Viral analysis, DNA, Viral genetics, Fanconi Anemia Complementation Group A Protein biosynthesis, Fanconi Anemia Complementation Group D2 Protein biosynthesis, Female, Gene Expression Profiling, Histones biosynthesis, Humans, Papillomavirus Infections metabolism, Papillomavirus Infections pathology, Papillomavirus Infections virology, Rad51 Recombinase biosynthesis, Tumor Suppressor Protein p53 biosynthesis, Vulva virology, Vulvar Diseases pathology, Vulvar Diseases virology, Alphapapillomavirus, Cell Cycle Checkpoints, DNA Damage, DNA Repair, Papillomavirus Infections genetics, Vulva pathology, Vulvar Diseases genetics
- Abstract
Human papillomavirus (HPV) infections may result in benign hyperplasia, caused by low-risk HPV types, or (pre)malignant lesions caused by high-risk HPV types. The molecular basis of this difference in malignant potential is not completely understood. Here, we performed gene profiling of different HPV infected vulvar tissues (condylomata acuminata (n = 5), usual type vulvar intraepithelial neoplasia (uVIN) (n = 9)) and control samples (n = 14) using Affymetrix Human U133A plus 2 GeneChips. Data were analyzed using OmniViz®, Partek® and Ingenuity® Software. Results were validated by real-time RT-PCR and immunostaining. Although similarities were observed between gene expression profiles of low- and high-risk HPV infected tissues (e.g., absence of estrogen receptor in condylomata and uVIN), high-risk HPV infected tissues showed more proliferation and displayed more DNA damage than tissues infected with low-risk HPV. These observations were confirmed by differential regulation of cell cycle checkpoints and by increased expression of DNA damage-biomarkers p53 and γH2AX. Furthermore, FANCA, FANCD2, BRCA1 and RAD51, key players in the DNA damage response, were significantly upregulated (p < 0.05). In addition, we compared our results with publicly available gene expression profiles of various other HPV-induced cancers (vulva, cervix and head-and-neck). This showed p16(INK4a) was the most significant marker to detect a high-risk HPV infection, but no other markers could be found. In conclusion, this study provides insight into the molecular basis of low- and high-risk HPV infections and indicates two main pathways (cell cycle and DNA damage response) that are much stronger affected by high-risk HPV as compared to low-risk HPV., (Copyright © 2011 UICC.)
- Published
- 2012
- Full Text
- View/download PDF
12. An autoimmune phenotype in vulvar lichen sclerosus and lichen planus: a Th1 response and high levels of microRNA-155.
- Author
-
Terlou A, Santegoets LA, van der Meijden WI, Heijmans-Antonissen C, Swagemakers SM, van der Spek PJ, Ewing PC, van Beurden M, Helmerhorst TJ, and Blok LJ
- Subjects
- Adult, Aged, Autoimmune Diseases metabolism, Autoimmune Diseases pathology, Cytokines biosynthesis, Cytokines genetics, Cytokines immunology, Dermis immunology, Dermis metabolism, Female, Gene Expression Profiling, Humans, Lichen Planus metabolism, Lichen Planus pathology, MicroRNAs biosynthesis, Middle Aged, T-Lymphocytes immunology, Vulvar Lichen Sclerosus metabolism, Vulvar Lichen Sclerosus pathology, Young Adult, Autoimmune Diseases immunology, Lichen Planus immunology, MicroRNAs immunology, Th1 Cells immunology, Vulvar Lichen Sclerosus immunology
- Abstract
Vulvar lichen sclerosus and lichen planus are T-cell-mediated chronic skin disorders. Although autoimmunity has been suggested, the exact pathogenesis of these disorders is still unknown. Therefore, the aim of the current study was to investigate the molecular and immunological mechanisms critical to the pathogenesis of vulvar lichen sclerosus and lichen planus. By using gene expression profiling and real-time RT-PCR experiments, we demonstrated a significantly increased expression of the pro-inflammatory cytokines (IFNγ, CXCR3, CXCL9, CXCL10, CXCL11, CCR5, CCL4, and CCL5) specific for a Th1 IFNγ-induced immune response. In addition, BIC/microRNA-155 (miR-155)--a microRNA involved in regulation of the immune response--was significantly upregulated in lichen sclerosus and lichen planus (9.5- and 17.7-fold change, respectively). Immunohistochemistry showed a significant T-cell response, with pronounced dermal infiltrates of CD4(+), CD8(+), and FOXP3(+) cells. In conclusion, these data demonstrate an autoimmune phenotype in vulvar lichen sclerosus and lichen planus, characterized by increased levels of Th1-specific cytokines, a dense T-cell infiltrate, and enhanced BIC/miR-155 expression.
- Published
- 2012
- Full Text
- View/download PDF
13. Progesterone inhibits epithelial-to-mesenchymal transition in endometrial cancer.
- Author
-
van der Horst PH, Wang Y, Vandenput I, Kühne LC, Ewing PC, van Ijcken WF, van der Zee M, Amant F, Burger CW, and Blok LJ
- Subjects
- Aged, Aged, 80 and over, Cell Line, Tumor, Cell Movement drug effects, Disease Progression, Endometrial Neoplasms genetics, Endometrial Neoplasms immunology, Endometrial Neoplasms metabolism, Female, Forkhead Transcription Factors metabolism, Genomics, Humans, Middle Aged, Receptors, Progesterone metabolism, Retrospective Studies, T-Lymphocyte Subsets drug effects, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Transcriptome drug effects, Endometrial Neoplasms pathology, Epithelial-Mesenchymal Transition drug effects, Progesterone pharmacology
- Abstract
Background: Every year approximately 74,000 women die of endometrial cancer, mainly due to recurrent or metastatic disease. The presence of tumor infiltrating lymphocytes (TILs) as well as progesterone receptor (PR) positivity has been correlated with improved prognosis. This study describes two mechanisms by which progesterone inhibits metastatic spread of endometrial cancer: by stimulating T-cell infiltration and by inhibiting epithelial-to-mesenchymal cell transition (EMT)., Methodology and Principal Findings: Paraffin sections from patients with (n = 9) or without (n = 9) progressive endometrial cancer (recurrent or metastatic disease) were assessed for the presence of CD4+ (helper), CD8+ (cytotoxic) and Foxp3+ (regulatory) T-lymphocytes and PR expression. Progressive disease was observed to be associated with significant loss of TILs and loss of PR expression. Frozen tumor samples, used for genome-wide expression analysis, showed significant regulation of pathways involved in immunesurveillance, EMT and metastasis. For a number of genes, such as CXCL14, DKK1, DKK4, PEG10 and WIF1, quantitive RT-PCR was performed to verify up- or downregulation in progressive disease. To corroborate the role of progesterone in regulating invasion, Ishikawa (IK) endometrial cancer cell lines stably transfected with PRA (IKPRA), PRB (IKPRB) and PRA+PRB (IKPRAB) were cultured in presence/absence of progesterone (MPA) and used for genome-wide expression analysis, Boyden- and wound healing migration assays, and IHC for known EMT markers. IKPRB and IKPRAB cell lines showed MPA induced inhibition of migration and loss of the mesenchymal marker vimentin at the invasive front of the wound healing assay. Furthermore, pathway analysis of significantly MPA regulated genes showed significant down regulation of important pathways involved in EMT, immunesuppression and metastasis: such as IL6-, TGF-β and Wnt/β-catenin signaling., Conclusion: Intact progesterone signaling in non-progressive endometrial cancer seems to be an important factor stimulating immunosurveilance and inhibiting transition from an epithelial to a more mesenchymal, more invasive phenotype.
- Published
- 2012
- Full Text
- View/download PDF
14. Identification of quiescent, stem-like cells in the distal female reproductive tract.
- Author
-
Wang Y, Sacchetti A, van Dijk MR, van der Zee M, van der Horst PH, Joosten R, Burger CW, Grootegoed JA, Blok LJ, and Fodde R
- Subjects
- Animals, Cell Separation methods, Cells, Cultured, Endometrium cytology, Endometrium physiology, Female, Humans, Mice, Mice, Transgenic, Cell Differentiation physiology, Oviducts cytology, Oviducts physiology, Stem Cells cytology, Stem Cells physiology
- Abstract
In fertile women, the endometrium undergoes regular cycles of tissue build-up and regression. It is likely that uterine stem cells are involved in this remarkable turn over. The main goal of our current investigations was to identify slow-cycling (quiescent) endometrial stem cells by means of a pulse-chase approach to selectively earmark, prospectively isolate, and characterize label-retaining cells (LRCs). To this aim, transgenic mice expressing histone2B-GFP (H2B-GFP) in a Tet-inducible fashion were administered doxycycline (pulse) which was thereafter withdrawn from the drinking water (chase). Over time, dividing cells progressively loose GFP signal whereas infrequently dividing cells retain H2B-GFP expression. We evaluated H2B-GFP retaining cells at different chase time points and identified long-term (LT; >12 weeks) LRCs. The LT-LRCs are negative for estrogen receptor-α and express low levels of progesterone receptors. LRCs sorted by FACS are able to form spheroids capable of self-renewal and differentiation. Upon serum stimulation spheroid cells are induced to differentiate and form glandular structures which express markers of mature műllerian epithelial cells. Overall, the results indicate that quiescent cells located in the distal oviduct have stem-like properties and can differentiate into distinct cell lineages specific of endometrium, proximal and distal oviduct. Future lineage-tracing studies will elucidate the role played by these cells in homeostasis, tissue injury and cancer of the female reproductive tract in the mouse and eventually in man.
- Published
- 2012
- Full Text
- View/download PDF
15. AMG 479, a novel IGF-1-R antibody, inhibits endometrial cancer cell proliferation through disruption of the PI3K/Akt and MAPK pathways.
- Author
-
Mendivil A, Zhou C, Cantrell LA, Gehrig PA, Malloy KM, Blok LJ, Burger CW, and Bae-Jump VL
- Subjects
- Antibodies, Monoclonal, Humanized, Apoptosis drug effects, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Endometrial Neoplasms enzymology, Endometrial Neoplasms metabolism, Endometrial Neoplasms pathology, Female, Humans, Phosphorylation, RNA, Messenger genetics, RNA, Messenger metabolism, Receptor, IGF Type 1 metabolism, Telomerase biosynthesis, Telomerase genetics, Antibodies, Monoclonal pharmacology, Endometrial Neoplasms drug therapy, MAP Kinase Signaling System drug effects, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Receptor, IGF Type 1 antagonists & inhibitors
- Abstract
Our goal was to evaluate the therapeutic potential of a novel antibody to the insulin growth factor-1 receptor (IGF-1-R; AMG 479) in endometrial cancer cells. The endometrial cancer cell lines, ECC-1/PRAB72 and RL-95-2, were used. Treatment with AMG 479 (0.02-200 nmol/L) resulted in inhibition of cell proliferation at 72 to 120 hours. Insulin growth factor-1 (0.15-7.5 nmol/L) stimulated growth in both cell lines (range of 15%-42%, P = .0025-.0445), which could be blocked by pretreatment with AMG 479 (mean of 29% for ECC-1/PRAB72, P = .006-.007; mean of 36% for RL-95-2, P = .0002-.0045). AMG 479 suppressed IGF-1-R kinase activity in a dose-dependent manner. Cells treated with AMG 479 underwent either G1 (ECC-1/PRAB72) or G2 (RL-95-2) arrest. AMG 479 decreased human telomerase reverse transcriptase (hTERT) mRNA expression in both endometrial cancer cell lines. Treatment with AMG 479 rapidly blocked IGF-1-induced phosphorylation of IFG-1-R, Akt, and p44/42. Thus, manipulation of the IGF-1-R pathway may serve as a promising therapeutic strategy for the treatment of endometrial cancer.
- Published
- 2011
- Full Text
- View/download PDF
16. Loss of APC function in mesenchymal cells surrounding the Müllerian duct leads to myometrial defects in adult mice.
- Author
-
Wang Y, Jia Y, Franken P, Smits R, Ewing PC, Lydon JP, DeMayo FJ, Burger CW, Anton Grootegoed J, Fodde R, and Blok LJ
- Subjects
- Adenomatous Polyposis Coli Protein genetics, Adenomatous Polyposis Coli Protein metabolism, Animals, Endometrium abnormalities, Endometrium metabolism, Endometrium pathology, Female, Gene Knockout Techniques, Genes, Reporter, Mice, Mice, Inbred C57BL, Myometrium metabolism, Myometrium pathology, Promoter Regions, Genetic, Receptors, Peptide genetics, Receptors, Transforming Growth Factor beta genetics, beta-Galactosidase biosynthesis, beta-Galactosidase genetics, Mesoderm pathology, Mullerian Ducts pathology, Myometrium abnormalities
- Abstract
The WNT signal transduction pathway plays a rate limiting role in early development of many different organs. To study the functional consequences of constitutive activation of the canonical WNT pathway in the developing uterus, we generated a novel mouse model where loss of the tumor suppressor gene Apc was induced. A mouse model was generated and evaluated where Amhr2(Cre/+) driven loss of Apc exon 15 was induced. The Apc recombination was detected mainly in the myometrial layer of the adult uterus. A significant loss of muscle fibers in myometrium was apparent, though with very few muscle cells earmarked by nuclear β-catenin. The finding was confirmed in the Pgr(Cre/+);Apc(15lox/15lox) mouse model. Loss of APC function in mesenchymal cells surrounding the fetal Müllerian ducts results in severe defects in the myometrial layers of the uterus in adult mice, suggesting that the WNT signaling pathway plays important roles in maintaining myometrial integrity., (Copyright © 2011 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
17. Progesterone receptor-B induction of BIRC3 protects endometrial cancer cells from AP1-59-mediated apoptosis.
- Author
-
Neubauer NL, Ward EC, Patel P, Lu Z, Lee I, Blok LJ, Hanifi-Moghaddam P, Schink J, and Kim JJ
- Subjects
- 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, Apoptosis drug effects, Ellipticines pharmacology, Endometrial Neoplasms metabolism, Gene Expression Regulation, Neoplastic genetics, Inhibitor of Apoptosis Proteins biosynthesis, Receptors, Progesterone metabolism
- 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., (© Springer Science+Business Media, LLC 2011)
- Published
- 2011
- Full Text
- View/download PDF
18. Nonsteroidal anti-inflammatory drugs do not interfere with imiquimod treatment for usual type vulvar intraepithelial neoplasia.
- Author
-
Terlou A, Kleinjan A, Beckmann I, Heijmans-Antonissen C, van Seters M, Santegoets LA, van Beurden M, Helmerhorst TJ, and Blok LJ
- Subjects
- Biopsy, Carcinoma in Situ immunology, Cell Separation, Drug Interactions, Female, Flow Cytometry, Fluorescent Dyes, Humans, Imiquimod, Immunohistochemistry, Vulvar Neoplasms immunology, Aminoquinolines therapeutic use, Anti-Inflammatory Agents, Non-Steroidal therapeutic use, Antineoplastic Agents therapeutic use, Carcinoma in Situ drug therapy, Vulvar Neoplasms drug therapy
- Abstract
Imiquimod has been shown to be an effective treatment for usual type vulvar intraepithelial neoplasia (uVIN). Since local inflammation and burning are common side effects, patients often use nonsteroidal anti-inflammatory drugs (NSAIDs). Our study investigated whether NSAID-use, which has been documented to inhibit the cell-mediated immune response, interferes with the outcome of imiquimod treatment. Monocyte-derived dendritic cells (moDCs) and Langerhans cells (moLCs) were cultured in the presence of NSAIDs. The expression of relevant surface markers (CD80, CD86, CD40, HLA-DR, CCR6 and CCR7), stimulatory function, and cytokine production were evaluated. Furthermore, we analyzed in uVIN patients whether frequent NSAID-use had an effect on the clinical response and on immunocompetent cell counts before and after imiquimod treatment. Although an effect was observed on the expression of moDC and moLC maturation markers, NSAIDs did not affect the ability of moDCs and moLCs to stimulate allogeneic T-cell proliferation, or the production of cytokines in an allogeneic T-cell stimulation assay. In agreement with this, in uVIN patients treated with imiquimod, no interference of frequent NSAID-use with clinical outcome was observed. However, we did notice that high CD1a(+) and CD207(+) cell counts in frequent NSAID-users before treatment seemed to predict a favourable response to imiquimod treatment. Our data indicate that NSAID-use does not seem to interfere with moDC and moLC function and does not interfere with immunomodulatory properties of imiquimod in uVIN patients. Therefore, NSAIDs can safely be used to reduce imiquimod side effects in uVIN patients during treatment., (Copyright © 2010 UICC.)
- Published
- 2011
- Full Text
- View/download PDF
19. Treatment of vulvar intraepithelial neoplasia with topical imiquimod: seven years median follow-up of a randomized clinical trial.
- Author
-
Terlou A, van Seters M, Ewing PC, Aaronson NK, Gundy CM, Heijmans-Antonissen C, Quint WG, Blok LJ, van Beurden M, and Helmerhorst TJ
- Subjects
- Adult, Body Image, Carcinoma in Situ pathology, Carcinoma in Situ psychology, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell pathology, Carcinoma, Squamous Cell psychology, Disease Progression, Double-Blind Method, Female, Follow-Up Studies, Humans, Imiquimod, Middle Aged, Neoplasm Invasiveness, Quality of Life, Sexuality, Vulvar Neoplasms pathology, Vulvar Neoplasms psychology, Aminoquinolines therapeutic use, Antineoplastic Agents therapeutic use, Carcinoma in Situ drug therapy, Vulvar Neoplasms drug therapy
- Abstract
Objective: Recently we reported on the efficacy of imiquimod for treating vulvar intraepithelial neoplasia (VIN) in a placebo-controlled, double-blinded randomized clinical trial (RCT). Four weeks after treatment, a complete response was observed in 35% of patients and a partial response in 46%. All complete responders remained disease-free at 12 months follow-up. In the current investigations, we assessed long-term follow-up at least 5 years after the initial RCT., Methods: Twenty-four of 26 imiquimod-treated patients who had participated in the initial RCT were seen for follow-up. Primary endpoint was durability of clinical response to imiquimod assessed by naked eye vulvar examination and histology. Long-term clinical response was correlated to lesion size before start of the initial RCT. Secondary endpoints were mental health, global quality of life, body image and sexual function in relation with long-term clinical response., Results: Median follow-up period was 7.2 years (range 5.6-8.3 years). VIN recurred in one of nine complete responders. Of the initial partial responders, two became disease-free after additional imiquimod treatment. In the other partial responders, VIN recurred at least once after the initial RCT. In long-term complete responders, lesion size at study entry was smaller and these patients had a significantly better global quality of life at follow-up than patients with residual disease and/or recurrence after imiquimod treatment., Conclusions: In case of a complete response, imiquimod is effective in the long-term. Furthermore, patients with a long-term complete response had a significantly better global quality of life than patients who recurred after imiquimod treatment., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
20. Imiquimod-induced clearance of HPV is associated with normalization of immune cell counts in usual type vulvar intraepithelial neoplasia.
- Author
-
Terlou A, van Seters M, Kleinjan A, Heijmans-Antonissen C, Santegoets LA, Beckmann I, van Beurden M, Helmerhorst TJ, and Blok LJ
- Subjects
- Adult, Biomarkers, Tumor metabolism, Carcinoma in Situ drug therapy, Carcinoma in Situ virology, Case-Control Studies, Cyclin-Dependent Kinase Inhibitor p16 metabolism, DNA, Viral genetics, Female, Humans, Imiquimod, Immunoenzyme Techniques, Middle Aged, Papillomavirus Infections drug therapy, Papillomavirus Infections virology, Polymerase Chain Reaction, Prognosis, Vulvar Neoplasms drug therapy, Vulvar Neoplasms virology, Young Adult, Aminoquinolines therapeutic use, Antineoplastic Agents therapeutic use, Carcinoma in Situ immunology, Lymphocyte Count, Papillomaviridae drug effects, Papillomavirus Infections immunology, Vulvar Neoplasms immunology
- Abstract
Recently, we reported on the efficacy of imiquimod for treatment of usual type vulvar intraepithelial neoplasia (uVIN). A histologic regression of uVIN to normal tissue was observed in 58% of patients. As success of treatment is related to clearance of high-risk human papilloma virus (HPV), the aim of our study was to assess differences in immune cell counts and in the expression of p16(INK4a) in VIN tissue before and after imiquimod treatment, in relation to HPV clearance and clinical response. Vulvar tissue samples taken prior to imiquimod treatment and 4 weeks after treatment were tested for the presence of HPV. Previously determined immune cell counts (CD1a, CD207, CD208, CD123/CD11c, CD94, CD4, CD8 and CD25/HLA-DR) in epidermis and dermis of 25 VIN patients and 19 healthy controls were completed with the counts for CD14 and CD68. The expression of p16(INK4a) was investigated by immunohistochemistry in 15 patients. Before imiquimod treatment, both HPV cleared and HPV noncleared patients showed mainly in the dermis significantly upregulated immune cell counts compared to healthy controls. However, in patients that cleared HPV and showed histologic regression already 4 weeks after imiquimod treatment, immune cell counts and p16(INK4a) expression were normalized. In conclusion, our data indicate that imiquimod-induced clearance of HPV results in normalization of counts for certain immune cells and is strongly correlated with histologic regression of the disease., (Copyright © 2010 UICC.)
- Published
- 2010
- Full Text
- View/download PDF
21. Wnt/Β-catenin and sex hormone signaling in endometrial homeostasis and cancer.
- Author
-
Wang Y, van der Zee M, Fodde R, and Blok LJ
- Subjects
- Animals, Carcinoma, Endometrioid genetics, Carcinoma, Endometrioid metabolism, Carcinoma, Endometrioid pathology, Endometrial Neoplasms genetics, Endometrial Neoplasms metabolism, Endometrial Neoplasms pathology, Endometrium drug effects, Endometrium metabolism, Endometrium pathology, Female, Gene Expression Regulation, Neoplastic drug effects, Gonadal Steroid Hormones metabolism, Gonadal Steroid Hormones pharmacology, Homeostasis drug effects, Homeostasis genetics, Homeostasis physiology, Humans, Models, Biological, Signal Transduction drug effects, Signal Transduction genetics, Signal Transduction physiology, Wnt Proteins genetics, Wnt Proteins metabolism, beta Catenin genetics, beta Catenin metabolism, Carcinoma, Endometrioid etiology, Endometrial Neoplasms etiology, Endometrium physiology, Gonadal Steroid Hormones physiology, Wnt Proteins physiology, beta Catenin physiology
- Abstract
A delicate balance between estrogen and progestagen signaling underlies proper functioning of the female reproductive tract and, in particular, the monthly re- and degenerative phases characteristic of the menstrual cycle. Here, we propose that the canonical Wnt/β-catenin signaling pathway may underlie this finely tuned hormonal equilibrium in endometrial homeostasis and, upon its constitutive activation, lead to neoplastic transformation of the endometrium. During the menstrual cycle, estradiol will enhance Wnt/β-catenin signaling in the proliferative phase, while progesterone inhibits Wnt/β-catenin signaling, thus restraining estrogens' proliferative actions, during the secretory phase. In case of enhanced or unopposed estrogen signaling, constitutive activation of Wnt/β-catenin signaling will trigger endometrial hyperplasia, which may develop further into endometrial cancer.
- Published
- 2010
- Full Text
- View/download PDF
22. Premalignant epithelial disorders of the vulva: squamous vulvar intraepithelial neoplasia, vulvar Paget's disease and melanoma in situ.
- Author
-
Terlou A, Blok LJ, Helmerhorst TJ, and van Beurden M
- Subjects
- Carcinoma in Situ epidemiology, Carcinoma in Situ therapy, Epithelium pathology, Female, Humans, Melanoma epidemiology, Melanoma therapy, Paget Disease, Extramammary epidemiology, Paget Disease, Extramammary therapy, Precancerous Conditions epidemiology, Precancerous Conditions therapy, Skin Neoplasms epidemiology, Skin Neoplasms therapy, Vulvar Neoplasms epidemiology, Vulvar Neoplasms therapy, Carcinoma in Situ diagnosis, Melanoma diagnosis, Paget Disease, Extramammary diagnosis, Precancerous Conditions diagnosis, Skin Neoplasms diagnosis, Vulvar Neoplasms diagnosis
- Abstract
No standard screening programs exist to detect vulvar carcinoma or its precursor lesions, and therefore gynecologists, dermatologists and other healthcare providers in this field should be aware of the clinical features, behavior and management of the different existing premalignant vulvar lesions, squamous vulvar intraepithelial neoplasia (VIN), vulvar Paget's disease and melanoma in situ. In 2004, a new classification for squamous VIN was introduced by the International Society for the Study of Vulvar Disease, subdividing squamous VIN into the HPV-related usual type, and into differentiated type, which is associated with lichen sclerosus. This review describes the relevant aspects of squamous VIN, vulvar Paget's disease and melanoma in situ, its epidemiological characteristics, diagnosis, management and malignant potential.
- Published
- 2010
- Full Text
- View/download PDF
23. Microsomal epoxide hydrolase expression in the endometrial uterine corpus is regulated by progesterone during the menstrual cycle.
- Author
-
Popp SL, Abele IS, Buck MB, Stope MB, Blok LJ, Hanifi-Moghaddam P, Burger CW, Fritz P, and Knabbe C
- Subjects
- 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, Endometrium enzymology, Epoxide Hydrolases biosynthesis, Gene Expression Regulation, Enzymologic, Menstrual Cycle physiology, Progesterone physiology
- 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.
- Published
- 2010
- Full Text
- View/download PDF
24. Progesterone inhibition of Wnt/beta-catenin signaling in normal endometrium and endometrial cancer.
- Author
-
Wang Y, Hanifi-Moghaddam P, Hanekamp EE, Kloosterboer HJ, Franken P, Veldscholte J, van Doorn HC, Ewing PC, Kim JJ, Grootegoed JA, Burger CW, Fodde R, and Blok LJ
- Subjects
- 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, Endometrial Neoplasms metabolism, Endometrium drug effects, Endometrium metabolism, Progesterone pharmacology, Signal Transduction drug effects, Wnt Proteins metabolism, beta Catenin metabolism
- 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.
- Published
- 2009
- Full Text
- View/download PDF
25. Genome-wide pathway analysis of folate-responsive genes to unravel the pathogenesis of orofacial clefting in man.
- Author
-
Bliek BJ, Steegers-Theunissen RP, Blok LJ, Santegoets LA, Lindemans J, Oostra BA, Steegers EA, and de Klein A
- Subjects
- Cell Line, Transformed, Child, Cleft Lip metabolism, Cleft Palate metabolism, Down-Regulation genetics, Female, Humans, Male, Pilot Projects, Up-Regulation genetics, Cleft Lip genetics, Cleft Palate genetics, Folic Acid physiology, Gene Expression Profiling, Gene Expression Regulation, Developmental physiology, Genome, Human physiology, Signal Transduction genetics
- Abstract
Background: A cleft of the lip with or without the palate (CLP) is a frequent congenital malformation with a heterogeneous etiology, for which folic acid supplementation has a protective effect. To gain more insight into the molecular pathways affected by natural folate, we examined gene expression profiles of cultured B-lymphoblasts from CLP patients before and after the addition of 5-methyltetrahydrofolate (5-mTHF) to the cultures., Methods: Immortalized B-lymphoblasts from five children with CLP were cultured in folate-deficient medium for 5 days. 5-mTHF was added to a concentration of 30 nM. Gene expression patterns were then evaluated before and after supplementation using Human Genome U133 Plus 2.0 arrays. Data analysis was performed with Omniviz and the GEPAS analysis suite. Differential genes were categorized into biological pathways with Ingenuity Pathway systems. Differential expression was validated by quantitative RT-PCR., Results: Using supervised clustering, with a false discovery rate <1%, we identified 144 and 409 significantly up-regulated and down-regulated probesets, respectively, after 5-mTHF addition. The regulated genes were involved in a variety of biological pathways, including one carbon pool and cell cycle regulation, biosynthesis of amino acids and DNA/RNA nucleotides, protein processing, apoptosis, and DNA repair., Conclusions: The large variety of the identified folate responsive pathways fits with the modifying role of folate via the methylation pathway. From the present data we may conclude that folate deficiency deranges normal cell development, which might contribute to the development of CLP. The role of these folate responsive genes in CLP development is intriguing and needs further investigation., ((c) 2008 Wiley-Liss, Inc.)
- Published
- 2008
- Full Text
- View/download PDF
26. Reduced local immunity in HPV-related VIN: expression of chemokines and involvement of immunocompetent cells.
- Author
-
Santegoets LA, van Seters M, Heijmans-Antonissen C, Kleinjan A, van Beurden M, Ewing PC, Kühne LC, Beckmann I, Burger CW, Helmerhorst TJ, and Blok LJ
- Subjects
- Adult, Chemokines genetics, Down-Regulation, Female, Gene Expression Regulation, Neoplastic, Humans, Immunohistochemistry, Microarray Analysis, Middle Aged, Papillomavirus Infections metabolism, Papillomavirus Infections virology, Reverse Transcriptase Polymerase Chain Reaction, Risk Factors, Tumor Virus Infections metabolism, Tumor Virus Infections virology, Up-Regulation, Uterine Cervical Neoplasms virology, Uterine Cervical Dysplasia virology, Alphapapillomavirus, Chemokines metabolism, Dendritic Cells immunology, Immunocompromised Host, Papillomavirus Infections complications, T-Lymphocytes immunology, Tumor Virus Infections complications, Uterine Cervical Neoplasms immunology, Uterine Cervical Dysplasia immunology
- Abstract
Usual type VIN is a premalignant disorder caused by persistent HPV infection. High prevalence of VIN in immuno-suppressed women suggests that a good innate and adaptive immune response is important for defense against HPV. Here, we explored expression levels of chemokines and related these to the presence or absence of immuno-competent cells (dendritic and T-cells) in affected (HPV-positive VIN) and non-affected (HPV-negative) vulvar tissues from the same patients. Combining microarray data with quantitative real-time RT-PCR, it was observed that several important chemokines were differentially expressed between VIN and control samples (up-regulation of IL8, CXCL10, CCL20 and CCL22 and down-regulation of CXCL12, CCL21 and CCL14). Furthermore, an increased number of mature dendritic cells (CD208+) seemed to be bottled up in the dermis, and although a T-cell response (increased CD4+ and CD8+ cells) was observed in VIN, a much larger response is required to clear the infection. In summary, it seems that most mature dendritic cells do not receive the proper chemokine signal for migration and will stay in the dermis, not able to present viral antigen to naive T-cells in the lymph node. Consequently the adaptive immune response diminishes, resulting in a persistent HPV infection with increased risk for neoplasia.
- Published
- 2008
- Full Text
- View/download PDF
27. The regulation and function of the forkhead transcription factor, Forkhead box O1, is dependent on the progesterone receptor in endometrial carcinoma.
- Author
-
Ward EC, Hoekstra AV, Blok LJ, Hanifi-Moghaddam P, Lurain JR, Singh DK, Buttin BM, Schink JC, and Kim JJ
- Subjects
- Cell Line, Endometrial Neoplasms drug therapy, Endometrial Neoplasms pathology, Endometrium chemistry, Female, Forkhead Box Protein O1, Forkhead Transcription Factors analysis, Humans, Phosphatidylinositol 3-Kinases physiology, Phosphorylation, Protein Processing, Post-Translational, Proto-Oncogene Proteins c-akt physiology, Receptors, Progesterone analysis, S-Phase Kinase-Associated Proteins physiology, Endometrial Neoplasms metabolism, Forkhead Transcription Factors physiology, Receptors, Progesterone physiology
- Abstract
In many type I endometrial cancers, the PTEN gene is inactivated, which ultimately leads to constitutively active Akt and the inhibition of Forkhead box O1 (FOXO1), a member of the FOXO subfamily of Forkhead/winged helix family of transcription factors. The expression, regulation, and function of FOXO1 in endometrial cancer were investigated in this study. Immunohistochemical analysis of 49 endometrial tumor tissues revealed a decrease of FOXO1 expression in 95.9% of the cases compared with the expression in normal endometrium. In four different endometrial cancer cell lines (ECC1, Hec1B, Ishikawa, and RL95), FOXO1 mRNA was expressed at similar levels; however, protein levels were low or undetectable in Ecc1, Ishikawa, and RL95 cells. Using small interfering RNA technology, we demonstrated that the low levels of FOXO1 protein were due to the involvement of Skp2, an oncogenic subunit of the Skp1/Cul1/F-box protein ubiquitin complex, given that silencing Skp2 increased FOXO1 protein expression in Ishikawa cells. Inhibition of Akt in Ishikawa cells also increased nuclear FOXO1 protein levels. Additionally, progestins increased FOXO1 protein levels, specifically through progesterone receptor B (PRB) as determined by using stably transfected PRA-specific and PRB-specific Ishikawa cell lines. Finally, overexpression of triple mutant (Tm) FOXO1 in the PR-specific Ishikawa cell lines caused cell cycle arrest and significantly decreased proliferation in the presence and absence of the progestin, R5020. Furthermore, TmFOXO1 overexpression induced apoptosis in PRB-specific cells in the presence and absence of ligand. Taken together, these data provide insight into the phosphoinositide-3-kinase/Akt/FOXO pathway for the determination of progestin responsiveness and the development of alternate therapies for endometrial cancer.
- Published
- 2008
- Full Text
- View/download PDF
28. Signaling by estrogens and tamoxifen in the human endometrium.
- Author
-
Gielen SC, Santegoets LA, Hanifi-Moghaddam P, Burger CW, and Blok LJ
- Subjects
- Cluster Analysis, Female, Gene Expression Profiling, Gene Expression Regulation, Humans, Polyps pathology, Endometrium drug effects, Endometrium metabolism, Estrogens metabolism, Signal Transduction drug effects, Tamoxifen pharmacology
- 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.
- Published
- 2008
- Full Text
- View/download PDF
29. Difference in signalling between various hormone therapies in endometrium, myometrium and upper part of the vagina.
- Author
-
Hanifi-Moghaddam P, Boers-Sijmons B, Klaassens AH, van Wijk FH, Van Ijcken WF, Van der Spek P, Verheul HA, Kloosterboer HJ, Burger CW, and Blok LJ
- Subjects
- Cluster Analysis, Drug Therapy, Combination, Estradiol therapeutic use, Female, Gene Expression drug effects, Gene Expression Profiling, Humans, Medroxyprogesterone Acetate therapeutic use, Norpregnenes therapeutic use, Endometrium metabolism, Estrogen Replacement Therapy, Myometrium metabolism, Signal Transduction drug effects, Vagina metabolism
- 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.
- Published
- 2008
- Full Text
- View/download PDF
30. Progesterone-mediated regulation of catechol-O-methyl transferase expression in endometrial cancer cells.
- Author
-
Salih SM, Salama SA, Jamaluddin M, Fadl AA, Blok LJ, Burger CW, Nagamani M, and Al-Hendy A
- Subjects
- Adenocarcinoma genetics, Catechol O-Methyltransferase biosynthesis, Endometrial Neoplasms genetics, Female, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Humans, Mutation, Protein Isoforms, Tumor Cells, Cultured, Adenocarcinoma metabolism, Catechol O-Methyltransferase genetics, Cell Proliferation drug effects, Endometrial Neoplasms metabolism, Progesterone metabolism, Receptors, Progesterone metabolism
- Abstract
The effects of estrogen and progesterone on the expression of estrogen-metabolizing enzymes such as catechol-O-methyl transferase (COMT) are not known. COMT converts genotoxic catecholestrogens to anticarcinogenic methoxyestrogens in the endometrium. The aim of this study is to investigate the effect of progesterone on COMT expression in well-differentiated endometrial cancer cells. The wild-type Ishikawa cell line as well as progesterone receptor A- or progesterone receptor B-transfected Ishikawa cells were used for in vitro studies. The regulation of COMT expression by progesterone was studied using Western blots, Hoechst dye DNA proliferation studies, and wild-type and/or site-directed mutagenesis of COMT promoter 1-luciferase reporter gene. Progesterone upregulated COMT protein expression in Ishikawa cells through progesterone receptor A isoform. COMT promoter activity was differentially regulated by the 3 half-site progesterone response elements in the COMT promoter. High doses of 2-ME2 inhibited Ishikawa cell proliferation. These data suggest that COMT expression is hormonally regulated in well-differentiated human endometrial cancer cells. COMT regulation and 2-ME2 production in the endometrium may affect endometrial carcinogenesis.
- Published
- 2008
- Full Text
- View/download PDF
31. Genomic and nongenomic effects of estrogen signaling in human endometrial cells: involvement of the growth factor receptor signaling downstream AKT pathway.
- Author
-
Gielen SC, Santegoets LA, Kühne LC, Van Ijcken WF, Boers-Sijmons B, Hanifi-Moghaddam P, Helmerhorst TJ, Blok LJ, and Burger CW
- Subjects
- 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, Endometrium metabolism, Estradiol pharmacology, Estrogens physiology, Proto-Oncogene Proteins c-akt metabolism, Receptors, Growth Factor metabolism, Tamoxifen pharmacology
- 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.
- Published
- 2007
- Full Text
- View/download PDF
32. HPV related VIN: highly proliferative and diminished responsiveness to extracellular signals.
- Author
-
Santegoets LA, Seters Mv, Helmerhorst TJ, Heijmans-Antonissen C, Hanifi-Moghaddam P, Ewing PC, van Ijcken WF, van der Spek PJ, van der Meijden WI, and Blok LJ
- Subjects
- 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, Alphapapillomavirus, Carcinoma in Situ virology, Precancerous Conditions virology, Signal Transduction, Vulvar Neoplasms virology
- 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.
- Published
- 2007
- Full Text
- View/download PDF
33. Molecular analysis of human endometrium: short-term tibolone signaling differs significantly from estrogen and estrogen + progestagen signaling.
- Author
-
Hanifi-Moghaddam P, Boers-Sijmons B, Klaassens AH, van Wijk FH, den Bakker MA, Ott MC, Shipley GL, Verheul HA, Kloosterboer HJ, Burger CW, and Blok LJ
- Subjects
- Cluster Analysis, Drug Therapy, Combination, Endometrium metabolism, Endometrium surgery, Female, Gene Expression drug effects, Gene Expression Profiling methods, Gene Regulatory Networks drug effects, Humans, Oligonucleotide Array Sequence Analysis, Polymerase Chain Reaction, Postmenopause, RNA, Messenger metabolism, Reproducibility of Results, Sex Hormone-Binding Globulin metabolism, Signal Transduction genetics, Uterine Prolapse metabolism, Uterine Prolapse surgery, Endometrium drug effects, Estradiol adverse effects, Estrogen Replacement Therapy adverse effects, Hysterectomy, Vaginal, Medroxyprogesterone adverse effects, Norpregnenes adverse effects, Signal Transduction drug effects, Uterine Prolapse drug therapy
- Abstract
Tibolone, a tissue-selective compound with a combination of estrogenic, progestagenic, and androgenic properties, is used as an alternative for estrogen or estrogen plus progesterone hormone therapy for the treatment of symptoms associated with menopause and osteoporosis. The current study compares the endometrial gene expression profiles after short-term (21 days) treatment with tibolone to the profiles after treatment with estradiol-only (E(2)) and E(2) + medroxyprogesterone acetate (E(2) + MPA) in healthy postmenopausal women undergoing hysterectomy for endometrial prolapse. The impact of E(2) treatment on endometrial gene expression (799 genes) was much higher than the effect of tibolone (173 genes) or E(2) + MPA treatment (174 genes). Furthermore, endometrial gene expression profiles after tibolone treatment show a weak similarity to the profiles after E(2) treatment (overlap 72 genes) and even less profile similarity to E(2) + MPA treatment (overlap 17 genes). Interestingly, 95 tibolone-specific genes were identified. Translation of profile similarity into biological processes and pathways showed that ER-mediated downstream processes, such as cell cycle and cell proliferation, are not affected by E2 + MPA, slightly by tibolone, but are significantly affected by E(2). In conclusion, tibolone treatment results in a tibolone-specific gene expression profile in the human endometrium, which shares only limited resemblance to E(2) and even less resemblance to E2 + MPA induced profiles.
- Published
- 2007
- Full Text
- View/download PDF
34. Levels of tibolone and estradiol and their nonsulfated and sulfated metabolites in serum, myometrium, and vagina of postmenopausal women following treatment for 21 days with tibolone, estradiol, or estradiol plus medroxyprogestrone acetate.
- Author
-
Verheul HA, Blok LJ, Burger CW, Hanifi-Moghaddam P, and Kloosterboer HJ
- Subjects
- 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, Estradiol metabolism, Estrone analogs & derivatives, Medroxyprogesterone Acetate metabolism, Myometrium metabolism, Norpregnenes metabolism, Selective Estrogen Receptor Modulators metabolism, Vagina metabolism
- 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.
- Published
- 2007
- Full Text
- View/download PDF
35. The hormone replacement therapy drug tibolone acts very similar to medroxyprogesterone acetate in an estrogen-and progesterone-responsive endometrial cancer cell line.
- Author
-
Hanifi-Moghaddam P, Sijmons B, Ott MC, van Ijcken WF, Nowzari D, Kuhne EC, van der Spek P, Kloosterboer HJ, Burger CW, and Blok LJ
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Endometrial Neoplasms metabolism, Estrogens pharmacology, Female, Gene Expression Profiling, Humans, Oligonucleotide Array Sequence Analysis, Progesterone pharmacology, Transcription, Genetic drug effects, Transcription, Genetic genetics, Endometrial Neoplasms genetics, Endometrial Neoplasms pathology, Estrogens metabolism, Gene Expression Regulation, Neoplastic drug effects, Hormone Replacement Therapy, Medroxyprogesterone Acetate pharmacology, Norpregnenes pharmacology, Progesterone metabolism
- Abstract
Tibolone, a steroidogenic compound with both estrogenic and progestagenic properties, is used as an alternative for estrogen or estrogen plus progesterone hormone therapy for the treatment of symptoms associated with menopause and osteoporosis. We have evaluated whether the effect of tibolone on a human endometrial cell line is similar to, or comparable with, the effect of estradiol (E(2)), medroxyprogesterone acetate (MPA) or E(2) + MPA treatment. Using stable transfection techniques, the estrogen receptor (ER) expressing human endometrial cancer cell line, ECC1, was altered to also express both progesterone receptors (PRs). These cells were then used to assess growth regulation and expression profiling (Affymetrix U133plus2) under the influence of E(2) (1 nM), MPA (1 nM), E(2) + MPA or tibolone (100 nM). Growth assessment and comparison of profiles indicate that tibolone behaves predominantly like MPA. Furthermore, regulation of prereplication complex genes, such as the minichromosome maintenance genes, could be involved in the observed strong inhibition of growth by tibolone as well as MPA. In addition, in total, 15 genes were found to be specific for tibolone treatment. These genes were predominantly involved in regulation of the cell cycle and differentiation.
- Published
- 2006
- Full Text
- View/download PDF
36. Ligand-controlled interaction of histone acetyltransferase binding to ORC-1 (HBO1) with the N-terminal transactivating domain of progesterone receptor induces steroid receptor coactivator 1-dependent coactivation of transcription.
- Author
-
Georgiakaki M, Chabbert-Buffet N, Dasen B, Meduri G, Wenk S, Rajhi L, Amazit L, Chauchereau A, Burger CW, Blok LJ, Milgrom E, Lombès M, Guiochon-Mantel A, and Loosfelt H
- Subjects
- Animals, Cell Line, Chlorocebus aethiops, Hormones metabolism, Humans, Ligands, Nuclear Receptor Coactivator 1, Origin Recognition Complex genetics, Protein Binding, Protein Subunits genetics, Protein Subunits metabolism, Receptors, Progesterone genetics, Receptors, Steroid metabolism, Signal Transduction, Transcription Factors, Histone Acetyltransferases metabolism, Origin Recognition Complex metabolism, Receptors, Progesterone metabolism, Transcription, Genetic genetics, Transcriptional Activation genetics
- Abstract
Modulators of cofactor recruitment by nuclear receptors are expected to play an important role in the coordination of hormone-induced transactivation processes. To identify such factors interacting with the N-terminal domain (NTD) of the progesterone receptor (PR), we used this domain as bait in the yeast Sos-Ras two-hybrid system. cDNAs encoding the C-terminal MYST (MOZ-Ybf2/Sas3-Sas2-Tip60 acetyltransferases) domain of HBO1 [histone acetyltransferase binding to the origin recognition complex (ORC) 1 subunit], a member of the MYST acetylase family, were thus selected from a human testis cDNA library. In transiently transfected CV1 cells, the wild-type HBO1 [611 amino acids (aa)] enhanced transcription mediated by steroid receptors, notably PR, mineralocorticoid receptor, and glucocorticoid receptor, and strongly induced PR and estrogen receptor coactivation by steroid receptor coactivator 1a (SRC-1a). As assessed by two-hybrid and glutathione-S-transferase pull-down assays, the HBO1 MYST acetylase domain (aa 340-611) interacts mainly with the NTD, and also contacts the DNA-binding domain and the hinge domains of hormone-bound PR. The HBO1 N-terminal region (aa 1-340) associates additionally with PR ligand-binding domain (LBD). HBO1 was found also to interact through its NTD with SRC-1a in the absence of steroid receptor. The latter coassociation enhanced specifically activation function 2 activation function encompassed in the LBD. Conversely, the MYST acetylase domain specifically enhanced SRC-1 coupling with PR NTD, through a hormone-dependent mechanism. In human embryonic kidney 293 cells expressing human PRA or PRB, HBO1 raised selectively an SRC-1-dependent response of PRB but failed to regulate PRA activity. We show that HBO1 acts through modification of an LBD-controlled structure present in the N terminus of PRB leading to the modulation of SRC-1 functional coupling with activation function 3-mediated transcription. Importantly, real-time RT-PCR analysis also revealed that HBO1 enhanced SRC-1 coactivation of PR-dependent transcription of human endogenous genes such as alpha-6 integrin and 11beta-hydroxydehydrogenase 2 but not that of amphiregulin. Immunofluorescence and confocal microscopy of human embryonic kidney-PRB cells demonstrated that the hormone induces the colocalization of HBO1 with PR-SRC-1 complex into nuclear speckles characteristic of PR-mediated chromatin remodeling. Our results suggest that HBO1 might play an important physiological role in human PR signaling.
- Published
- 2006
- Full Text
- View/download PDF
37. Histological and immunohistochemical evaluation of postmenopausal endometrium after 3 weeks of treatment with tibolone, estrogen only, or estrogen plus progestagen.
- Author
-
Klaassens AH, van Wijk FH, Hanifi-Moghaddam P, Sijmons B, Ewing PC, Ten Kate-Booij MJ, Kooi GS, Kloosterboer HJ, Blok LJ, and Burger CW
- Subjects
- Aged, Drug Synergism, Female, Humans, Hysterectomy, Immunohistochemistry, Middle Aged, Mitosis drug effects, Uterine Prolapse metabolism, Uterine Prolapse pathology, Uterine Prolapse surgery, Cell Proliferation drug effects, Endometrium metabolism, Endometrium pathology, Estrogens pharmacology, Norpregnenes pharmacology, Postmenopause metabolism, Progestins pharmacology
- 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.
- Published
- 2006
- Full Text
- View/download PDF
38. Growth regulation and transcriptional activities of estrogen and progesterone in human endometrial cancer cells.
- Author
-
Gielen SC, Hanekamp EE, Hanifi-Moghaddam P, Sijbers AM, van Gool AJ, Burger CW, Blok LJ, and Huikeshoven FJ
- Subjects
- Blotting, Western, Cell Differentiation genetics, Cell Line, Tumor, Cell Proliferation, Estrogens metabolism, Female, Gene Expression Regulation, Neoplastic, Humans, Oligonucleotide Array Sequence Analysis, Progesterone metabolism, Receptors, Progesterone genetics, Sensitivity and Specificity, Endometrial Neoplasms pathology, Estrogens pharmacology, Progesterone pharmacology, Receptors, Progesterone metabolism
- Abstract
Estrogen-stimulated growth of the malignant human endometrium can be balanced by the differentiating properties of progesterone. To study the molecular basis behind this, gene expression profiling was performed using complementary DNA microarray analysis. In this study, the human endometrial cancer cell lines ECC-1 and PRAB-36 were used as models. The ECC-1 cell line, which expresses high levels of estrogen receptor alpha and is stimulated in growth by estrogens, was used to study estrogen regulation of gene expression. The Ishikawa sub-cell line PRAB-36, expressing both PRA and PRB, progesterone receptor isoforms, and inhibited in growth by progestagens, was used to study progesterone regulation of gene expression. Using these two well-differentiated human endometrial cancer cell lines, 148 estrogen- and 148 progesterone-regulated genes were identified. After functional classification, the estrogen- and progesterone-regulated genes could be categorized in different biologically relevant groups. Within the group of "cell growth and/or maintenance," 81 genes were clustered, from which a number of genes could be involved in arranging the cross talk that exists between estrogen and progesterone signaling. On the basis of analysis of the current findings, it is hypothesized that cross talk between estrogen and progestagen signaling does not occur by counterregulation of single genes, but rather at the level of differential regulation of different genes within the same functional families.
- Published
- 2006
- Full Text
- View/download PDF
39. Tamoxifen treatment for breast cancer enforces a distinct gene-expression profile on the human endometrium: an exploratory study.
- Author
-
Gielen SC, Kühne LC, Ewing PC, Blok LJ, and Burger CW
- Subjects
- Antineoplastic Agents, Hormonal pharmacology, Cell Proliferation drug effects, Endometrium drug effects, Endometrium pathology, ErbB Receptors genetics, Female, Gene Expression Profiling, Genes, Neoplasm, Genes, myc genetics, Genes, p53 genetics, Humans, Middle Aged, Tamoxifen pharmacology, Transcription Factor RelA genetics, beta Catenin genetics, Antineoplastic Agents, Hormonal therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Endometrium metabolism, Gene Expression drug effects, Tamoxifen therapeutic use
- Abstract
Tamoxifen treatment for breast cancer increases proliferation of the endometrium, resulting in an enhanced prevalence of endometrial pathologies, including endometrial cancer. An exploratory study was performed to begin to understand the molecular mechanism of tamoxifen action in the endometrium. Gene-expression profiles were generated of endometrial samples of tamoxifen users and compared with matched controls. The pathological classification of samples from both groups included atrophic/inactive endometrium and endometrial polyps. Unsupervised clustering revealed that samples of tamoxifen users were, irrespective of pathological classification, fairly similar and consequently form a subgroup distinct from the matched controls. Using SAM analysis (a statistical method to select genes differentially expressed between groups), 256 differentially expressed genes were selected between the tamoxifen and control groups. Upon comparing these genes with oestrogen-regulated genes, identified under similar circumstances, 95% of the differentially expressed genes turned out to be tamoxifen-specific. Finally, construction of a gene-expression network of the differentially expressed genes revealed that 69 genes centred around five well-known genes: TP53, RELA, MYC, epidermal growth factor receptor and beta-catenin. This could indicate that these well-known genes, and the pathways in which they function, are important for tamoxifen-controlled proliferation of the endometrium.
- Published
- 2005
- Full Text
- View/download PDF
40. Analysis of estrogen agonism and antagonism of tamoxifen, raloxifene, and ICI182780 in endometrial cancer cells: a putative role for the epidermal growth factor receptor ligand amphiregulin.
- Author
-
Gielen SC, Burger CW, Kühne LC, Hanifi-Moghaddam P, and Blok LJ
- Subjects
- Amphiregulin, EGF Family of Proteins, Estradiol pharmacology, Estradiol physiology, Female, Fulvestrant, Gene Expression Profiling, Humans, Transcription, Genetic, Tumor Cells, Cultured, 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
- 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.
- Published
- 2005
- Full Text
- View/download PDF
41. Differences in invasive capacity of endometrial cancer cell lines expressing different progesterone receptor isotypes: possible involvement of cadherins.
- Author
-
Hanekamp EE, Gielen SC, De Ruiter PE, Chadha-Ajwani S, Huikeshoven FJ, Burger CW, Grootegoed JA, and Blok LJ
- Subjects
- Blotting, Western, Cadherins biosynthesis, Female, Gene Expression Profiling, Humans, Immunohistochemistry, Neoplasm Metastasis, Oligonucleotide Array Sequence Analysis, Phenotype, Tumor Cells, Cultured, Endometrial Neoplasms pathology, Neoplasm Invasiveness genetics, Neoplasm Invasiveness physiopathology, Receptors, Progesterone biosynthesis
- Abstract
Objective: Loss of expression of progesterone receptors (PR) in endometrial cancer is related to a more invasive and metastatic phenotype. In this study we aim to investigate whether selective loss of PRA or PRB affects the invasive capacity of endometrial cancer cells., Methods: cDNA microarrays were performed to compare gene expression profiles of a set of endometrial cancer sub-cell lines expressing PRA and/or PRB. In vitro invasion assays were performed to assess whether differences in gene expression between the lines were reflected by their invasive behavior., Results: It was observed that cell lines that express only PRA express higher levels of cadherins, and show a lower level of invasion compared to cell lines that express PRB. When cadherin function was inhibited in exclusively PRA-expressing cell lines, an increase of in vitro invasion was observed. In support of these findings, it was observed that in higher grade and more invasive endometrial cancer, expression of E-cadherin decreased., Conclusions: These results indicate that relative loss of PRA during progression of endometrial cancer can have a negative impact on cadherin expression, which may lead to development of a more metastatic phenotype.
- Published
- 2005
- Full Text
- View/download PDF
42. Progesterone-induced inhibition of growth and differential regulation of gene expression in PRA- and/or PRB-expressing endometrial cancer cell lines.
- Author
-
Smid-Koopman E, Kuhne LC, Hanekamp EE, Gielen SC, De Ruiter PE, Grootegoed JA, Helmerhorst TJ, Burger CW, Brinkmann AO, Huikeshoven FJ, and Blok LJ
- Subjects
- Apoptosis, Cell Differentiation, Cell Proliferation, Female, Gene Expression Profiling, Humans, Protein Isoforms, Transfection, Tumor Cells, Cultured, Endometrial Neoplasms genetics, Endometrial Neoplasms pathology, Progesterone physiology, Receptors, Progesterone biosynthesis
- Abstract
Objective: Progesterone plays an important role in controlling proliferation and differentiation of the human endometrium. Because there are two progesterone receptor isoforms (PRA and PRB), it was important to generate tools to be able to study the role of these two progesterone receptors separately., Methods: Using stable transfection techniques, both human progesterone receptor isoforms (hPRA and hPRB) were reintroduced into a hPR-negative subclone of the well-differentiated endometrial cancer cell line Ishikawa. Several Ishikawa subcell lines were constructed, each expressing different levels of hPRA, hPRB, or hPRA and hPRB, respectively., Results: These Ishikawa subcell lines showed a marked progesterone-induced growth inhibition with induction of apoptosis after long-term culture in the presence of hormone. Upon measuring gene regulation, a clear difference in regulation of expression of the selected genes by progesterone treatment was observed between the PRA-, PRB-, or PRA/B-expressing cell lines. Integrin beta4 (ITGB4) was only regulated in PRA-expressing cells; amphiregulin was highly regulated in PRB-expressing cells; insulin-like growth factor binding protein 3 (IGFBP3) was only regulated in PRB- and PRA/B-expressing cells; and metallothionein 1L (MT1L) was highly regulated in PRA/B-expressing cells. Interestingly, based on literature data, these genes can be implicated in induction of apoptosis, but are modulated here in such a way that suggests induction of resistance against apoptosis., Conclusion: Reintroduction of PRs into Ishikawa cells rescued progesterone responsiveness in these cells. Furthermore, using these human endometrial cancer subcell lines, clear and distinct functional differences between the PR isoforms were observed.
- Published
- 2005
- Full Text
- View/download PDF
43. Expression profiling of androgen-dependent and -independent LNCaP cells: EGF versus androgen signalling.
- Author
-
Oosterhoff JK, Grootegoed JA, and Blok LJ
- Subjects
- Gene Expression Profiling, Humans, Male, Neoplasms, Hormone-Dependent genetics, Oligonucleotide Array Sequence Analysis, Prostatic Neoplasms genetics, Tumor Cells, Cultured, Androgens pharmacology, Biomarkers, Tumor metabolism, Epidermal Growth Factor pharmacology, Gene Expression Regulation, Neoplastic drug effects, Neoplasms, Hormone-Dependent metabolism, Prostatic Neoplasms metabolism, Signal Transduction
- Abstract
Prostate cancer development often includes a shift from androgen-dependent to androgen-independent growth. It is hypothesized that, during this transition, growth factors like the epidermal growth factor (EGF) gain importance as activators of tumour cell proliferation. To study this, androgen- and EGF-regulation of growth and gene-expression was analysed in the androgen-dependent human prostate cancer cell line LNCaP-FGC (FGC) and its androgen-independent derivative line LNCaP-LNO (LNO). It was observed that androgen-dependent FGC cells require exposure to either androgens or EGF to proliferate. This is in contrast to androgen-independent LNO cells that showed significant proliferation in medium depleted of androgens and growth factors. Gene expression data were obtained for the androgen-dependent FGC and androgen-independent LNO cells cultured in the presence or absence of androgens (synthetic R1881) or EGF for different time periods. Expression profiling showed that many cell cycle genes, including a number of androgen- and EGF-regulated genes, are constitutively activated in androgen-independent LNO cells. Furthermore, the overlap between changes in gene expression activated by androgen and EGF receptor signalling pathways was found to be very high (75%). These results partly explain why androgen-independent LNO cells can proliferate in the absence of androgenic stimulation. However, possibly other, so far unknown, signal transduction pathways that induce and maintain proliferation, have also been activated.
- Published
- 2005
- Full Text
- View/download PDF
44. EGF signalling in prostate cancer cell lines is inhibited by a high expression level of the endocytosis protein REPS2.
- Author
-
Oosterhoff JK, Kühne LC, Grootegoed JA, and Blok LJ
- Subjects
- Androgens pharmacology, Calcium-Binding Proteins, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Disease Progression, Early Growth Response Protein 1, Epidermal Growth Factor metabolism, ErbB Receptors genetics, ErbB Receptors metabolism, Gene Expression Regulation, Neoplastic, Humans, Immediate-Early Proteins genetics, Immediate-Early Proteins metabolism, Intracellular Signaling Peptides and Proteins genetics, Male, Neoplasms, Hormone-Dependent genetics, Prostatic Neoplasms genetics, Proto-Oncogene Proteins c-fos genetics, Proto-Oncogene Proteins c-fos metabolism, Proto-Oncogene Proteins c-jun genetics, Proto-Oncogene Proteins c-jun metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Signal Transduction, Transcription Factors genetics, Transcription Factors metabolism, Tumor Cells, Cultured, Endocytosis, Epidermal Growth Factor pharmacology, Intracellular Signaling Peptides and Proteins metabolism, Neoplasms, Hormone-Dependent metabolism, Prostatic Neoplasms metabolism
- Abstract
In advanced prostate cancer, cellular changes occur leading to a transition from androgen-dependent to androgen-independent growth. During this transition, proliferation of androgen-dependent prostate cancer cells becomes more and more dependent on growth factors, like the epidermal growth factor (EGF). Endocytosis of growth factor receptors, one of the mechanisms that controls growth factor signalling, was observed to be markedly changed in advanced metastatic prostate cancer. Internalisation and signalling of EGF receptors was examined in different prostate cancer cell lines, in relation to the expression level of the endocytosis-related REPS2 gene. It was observed that a high level of REPS2 correlates with reduced EGF-internalisation. To investigate this more thoroughly, prostate cancer cells with inducible REPS2 expression were generated. Using these cells, it was found that REPS2-induction indeed results in reduction of EGF-internalisation. Furthermore, when EGF receptor signalling was evaluated, by examination of mRNA expression for several EGF-responsive genes (EGF receptor, EGR-1, Fos and Jun), it was observed that induced expression of REPS2 exerts an inhibiting effect on this signalling. From these experiments, it is concluded that increased REPS2 expression negatively affects EGF receptor internalisation and subsequent signalling. Therefore, decreased REPS2 expression during prostate cancer progression, observed in earlier work, may result in enhanced EGF receptor expression and signalling, which could add to the androgen-independent state of advanced prostate cancer.
- Published
- 2005
- Full Text
- View/download PDF
45. Molecular portrait of the progestagenic and estrogenic actions of tibolone: behavior of cellular networks in response to tibolone.
- Author
-
Hanifi-Moghaddam P, Gielen SC, Kloosterboer HJ, De Gooyer ME, Sijbers AM, van Gool AJ, Smid M, Moorhouse M, van Wijk FH, Burger CW, and Blok LJ
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma pathology, Cell Differentiation drug effects, Cell Division drug effects, Cell Line, Tumor, Endometrial Neoplasms genetics, Endometrial Neoplasms pathology, Estradiol pharmacology, Female, Gene Expression Profiling, Humans, Nerve Net, Transcription, Genetic drug effects, Estrogen Receptor Modulators pharmacology, Gene Expression Regulation, Neoplastic drug effects, Norpregnenes pharmacology, Progestins antagonists & inhibitors
- Abstract
Tibolone is a synthetic steroid with estrogenic effects on brain, vagina, and bone without stimulating the endometrium. During tibolone treatment, it is thought that the progestagenic properties of tibolone stimulate cell differentiation, which effectively counterbalances the growth-stimulating effects of the estrogenic properties of tibolone. The objective of this study was to characterize the expression profile that reflects the endometrial responses to the separated estrogenic (growth-inducing) and progestagenic (growth-inhibiting) actions of tibolone, thus gaining insight into the counteracting effect of these properties of tibolone on the endometrium. The estrogenic action of tibolone was studied in the estrogen-responsive ECC1 cell line (expressing estrogen receptor alpha), and the progestagenic action was studied in the progesterone-responsive cell line Ishikawa PRAB-36 (expressing PRA and PRB). The data showed that the progestagenic and estrogenic effects of tibolone produce different expression profiles with a narrow overlap in genes; however, both properties modulate the same biological processes. The final genetic network analysis indicated that the estrogenic effect of tibolone is potentially counterbalanced by the progestagenic metabolite of tibolone via differential regulation of similar cellular processes. For example, both progestagenic and estrogenic properties stimulate proliferation, but they exert the opposite effect on apoptosis. The apoptosis network was stimulated by the progestagenic properties of tibolone; in contrast, the estrogenic effect of tibolone suppressed the apoptosis network. The current results indicate that this differential regulation is realized through modulation of a different group of genes and rarely via contraregulation of the same set of genes.
- Published
- 2005
- Full Text
- View/download PDF
46. Steroid-modulated proliferation of human endometrial carcinoma cell lines: any role for insulin-like growth factor signaling?
- Author
-
Gielen SC, Hanekamp EE, Blok LJ, Huikeshoven FJ, and Burger CW
- Subjects
- Female, Humans, Insulin-Like Growth Factor Binding Protein 4 biosynthesis, Signal Transduction, Tumor Cells, Cultured, Up-Regulation, Carcinoma pathology, Cell Proliferation, Endometrial Neoplasms pathology, Estrogens pharmacology, Somatomedins pharmacology
- Abstract
Objectives: Estrogen-stimulated proliferation of the normal and malignant human endometrium is balanced by the differentiating properties of progesterone. This study evaluated the role of insulin-like growth factor (IGF) signaling in steroid-induced modulation of endometrial cancer cell proliferation., Methods: We used the human endometrial, estrogen-responsive ECC-1 and progesterone-responsive PRAB-36 cell lines. Proliferation studies with IGFs in combination with either estrogen or progesterone were conducted. Furthermore, the mRNA and protein expression of insulin-like growth factor-binding proteins (IGFBPs) was evaluated., Results: Using the ECC-1 cell line, we observed that estrogen-induced proliferation is modulated via the IGF-receptor signaling pathway, and that IGF-1-induced stimulation of proliferation does not influence estrogen receptor signaling. Furthermore, expression of the main modulators of IGF action, the IGFBPs, was found to be regulated by estrogen and progesterone in both cell lines. IGFBP-4 was up-regulated by estrogen in the ECC-1 cell line, and IGFBP-3 and IGFBP-6 were down-regulated by progesterone in the PRAB-36 cell line., Conclusion: Estrogen-induced stimulation of proliferation of ECC-1 endometrial cancer cells is partly achieved via IGF signaling. Furthermore, the IGFBPs are regulated by estrogens as well as progestagens and could potentially play a role in the modulation of endometrial cancer cell proliferation.
- Published
- 2005
- Full Text
- View/download PDF
47. Progesterone receptor A and B expression and progestagen treatment in growth and spread of endometrial cancer cells in nude mice.
- Author
-
Hanekamp EE, Kühne LM, Grootegoed JA, Burger CW, and Blok LJ
- Subjects
- Animals, Cell Line, Female, Gene Expression Regulation, Neoplastic, Medroxyprogesterone Acetate pharmacology, Mice, Mice, Nude, Neoplasm Invasiveness, Progestins pharmacology, Receptors, Progesterone genetics, Tumor Burden, Endometrial Neoplasms metabolism, Endometrial Neoplasms pathology, Receptors, Progesterone metabolism
- Abstract
In endometrial cancer, decreased expression of progesterone receptor (PR) isotypes A and B (PRA and PRB) is a feature of poorly differentiated tumours. In distant metastases, PRB is the predominantly expressed isotype and endometrial cancer cells that express PRB have been observed to be more invasive. Furthermore, PRB-associated in vitro invasion is markedly inhibited by progestagens. In the present study, ovariectomized mice were injected intraperitoneally with Ishikawa endometrial cancer cells that express only PRA, only PRB, both PRA and PRB, or no PR. Half of the mice were substituted with medroxyprogesterone acetate (MPA). After ten weeks, growth and spread of the cancer cells were examined macroscopically, microscopically, and by PCR detection. Without MPA substitution, cells that express only PRB were found to be the most proliferative and migrative, while cells that express only PRA, both receptor isotypes, or no PR, showed minimal growth and spread. MPA appeared to inhibit growth and spread of PR-positive cells. Surprisingly, when mice that were inoculated with PR-negative cells were substituted with MPA, this resulted in massive abdominal tumour growth. These results provide further evidence that over-expression of PRB in endometrial cancer contributes to the development of a more aggressive phenotype. MPA inhibits tumour growth and spread of PR-positive cells, but can also have an indirectly stimulating effect on PR-negative tumour cells, probably through a host-mediated response.
- Published
- 2004
- Full Text
- View/download PDF
48. Phosphorylation of androgen receptor isoforms.
- Author
-
Wong HY, Burghoorn JA, Van Leeuwen M, De Ruiter PE, Schippers E, Blok LJ, Li KW, Dekker HL, De Jong L, Trapman J, Grootegoed JA, and Brinkmann AO
- Subjects
- Animals, COS Cells, Cell Line, Chromatography, High Pressure Liquid, Electrophoresis, Polyacrylamide Gel, Humans, Mass Spectrometry, Metribolone pharmacology, Molecular Weight, Mutagenesis, Site-Directed, Mutation, Phosphorylation drug effects, Phosphoserine metabolism, Protein Isoforms chemistry, Protein Isoforms genetics, Protein Isoforms metabolism, Receptors, Androgen genetics, Transcription, Genetic, Receptors, Androgen chemistry, Receptors, Androgen metabolism
- Abstract
Phosphorylation of the human AR (androgen receptor) is directly correlated with the appearance of at least three AR isoforms on an SDS/polyacrylamide gel. However, it is still not clear to what extent phosphorylation is involved in the occurrence of isoforms, which sites are phosphorylated and what are the functions of these phosphosites. The human AR was expressed in COS-1 cells and AR phosphorylation was studied further by mutational analyses and by using reversed-phase HPLC and MS. The reversed-phase HPLC elution pattern of the three isoforms revealed that Ser-650 was phosphorylated constitutively. After de novo synthesis, only Ser-650 was phosphorylated in the smallest isoform of 110 kDa and both Ser-650 and Ser-94 were phosphorylated in the second isoform of 112 kDa. The hormone-induced 114 kDa isoform shows an overall increase in phosphorylation of all the isolated peptides. The activities of the Ser-Ala substitution mutant S650A (Ser-650-->Ala) was found to be identical with wild-type AR activation in four different cell lines and three different functional analyses, e.g. transactivation, N- and C-terminal-domain interaction and co-activation by transcriptional intermediary factor 2. This was also found for mutants S94A and S515A with respect to transactivation. However, the S515A mutation, which should eliminate phosphorylation of the potential mitogen-activated protein kinase site, Ser-515, resulted in an unphosphorylated form of the peptide containing Ser-650. This suggests that Ser-515 can modulate phosphorylation at another site. The present study shows that the AR isoform pattern from AR de novo synthesis is directly linked to differential phosphorylation of a distinct set of sites. After mutagenesis of these sites, no major change in functional activity of the AR was observed.
- Published
- 2004
- Full Text
- View/download PDF
49. Identification of REPS2 as a putative modulator of NF-kappaB activity in prostate cancer cells.
- Author
-
Penninkhof F, Grootegoed JA, and Blok LJ
- Subjects
- Animals, COS Cells, Calcium-Binding Proteins, Chlorocebus aethiops, Humans, Male, Tetradecanoylphorbol Acetate pharmacology, Transcription Factor RelA metabolism, Tryptophan physiology, Intracellular Signaling Peptides and Proteins, NF-kappa B metabolism, Prostatic Neoplasms enzymology, Proteins physiology
- Abstract
The protein REPS2 is implicated in growth factor receptor-mediated endocytosis and signalling, and its expression is downregulated in androgen-independent prostate cancer cells. Herein, the NF-kappaB subunit p65 is identified as a human REPS2 protein partner, interacting with the EH domain of REPS2. Using crystal structure data from literature and experimental data from yeast and mammalian two-hybrid analysis, the results indicate that the NPF-motif in p65 acts as binding site for the EH domain in REPS2. However, in cultured prostate cancer cells, the REPS2-p65 interaction is triggered upon stimulation with phorbol ester (PMA). This indicates that PMA-sensitive signalling pathways can affect the interaction between REPS2 and p65. During prostate cancer progression from androgen-dependent to androgen-independent growth, downregulation of REPS2 is accompanied by upregulation of NF-kappaB activity. This might involve loss of REPS2-p65 interaction, which would lead to increased NF-kappaB activity. Androgen-deprivation causes apoptosis of prostate cancer cells, and activated NF-kappaB is a known inhibitor of apoptosis. Hence, decreased expression of REPS2 might be a key factor, causing prostate cancer cells to become resistant to induction of apoptosis by androgen deprivation., (Copyright 2004 Nature Publishing Group)
- Published
- 2004
- Full Text
- View/download PDF
50. Gene expression profiling in human endometrial cancer tissue samples: utility and diagnostic value.
- Author
-
Smid-Koopman E, Blok LJ, Helmerhorst TJ, Chadha-Ajwani S, Burger CW, Brinkmann AO, and Huikeshoven FJ
- Subjects
- Adult, Aged, Aged, 80 and over, Endometrial Neoplasms diagnosis, Endometrial Neoplasms metabolism, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Middle Aged, Multigene Family, Oligonucleotide Array Sequence Analysis, Prognosis, Sensitivity and Specificity, Endometrial Neoplasms genetics
- Abstract
Objective: Recently, gene expression profiling techniques have been used on several human cancers to classify tumor subgroups with a specific biological behavior, which were previously undetected by the conventional histopathologic staging systems. In the current study, the clinical usefulness and prognostic value of gene expression profiling in human endometrial carcinomas were studied., Methods: A macro cDNA array, containing cDNAs of 588 genes selected from different areas of cancer research, was used to generate gene expression profiles of tumor tissue samples. The gene expression profiles of 12 endometrial cancers, 3 benign (e.g. noncancer) endometrial tissue samples and 3 myometrial tissue samples, taken from human surgical specimen, were compared., Results: The efficacy to generate a gene expression profile of these tissue samples was 77%. The RNA samples could be randomly taken from the tissue samples and were highly reproducible. Cluster analysis of gene expression profiles of the different samples showed that the benign endometrial and the myometrial samples clustered separately from the tumor samples, indicating that the gene expression profiles were tissue specific and not patient specific. Cluster analysis of the tumor samples revealed two distinct tumor clusters. Ranking of the tumors in the two clusters showed high similarity with the histopathologic classification [International Federation of Gynecology and Obstetrics (FIGO) grading]., Conclusion: Classification of endometrial tumors on basis of their gene expression profiles showed similarity with the FIGO grading system.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.