epidermal-growth-factor has been researched along with Endometrial-Neoplasms* in 70 studies
1 review(s) available for epidermal-growth-factor and Endometrial-Neoplasms
Article | Year |
---|---|
Mechanisms of growth inhibition by antiestrogens and progestins in human breast and endometrial cancer cells.
Marked changes in both growth factor and proto-oncogene expression occur due to treatment of hormonally-responsive human cancers with progestins and antiestrogens. In human endometrial cancer cell lines the antiproliferative effects of progestins and antiestrogens in a particular cell line appear to be associated with similar effects on growth factor and/or proto-oncogene expression. This suggests that although these compounds initially interact with different steroid hormone receptors, the molecular mechanisms of their growth inhibition may be essentially similar. In the case of human breast cancer cell lines, however, the effects of progestins and antiestrogens on gene regulation are often different, suggesting that the molecular mechanisms of progestin and antiestrogen growth inhibition may be essentially dissimilar. Topics: Breast Neoplasms; Cell Division; Endometrial Neoplasms; Epidermal Growth Factor; Estrogen Antagonists; Female; Gene Expression Regulation, Neoplastic; Humans; Progestins; Proto-Oncogene Mas; Proto-Oncogenes; Transforming Growth Factors | 1992 |
1 trial(s) available for epidermal-growth-factor and Endometrial-Neoplasms
Article | Year |
---|---|
[Concentration of epidermal growth factor (EGF) in serum of women with endometrial cancer].
Serum Epidermal Growth Factor (EGF) concentrations were estimated in 40 women with endometrial carcinoma and in 10 women with proliferative endometrium. Medium EGF level in study group was 0.97 +/- 0.41 ng/ml whereas 0.78 +/- 0.19 ng/ml in control group. The highest values of EGF (1.39 +/- 0.44 ng/ml) were found in moderately differentiated adenocarcinomas (G2) whereas the low values (0.65 +/- 0.18 ng/ml) in poorly differentiated adenocarcinomas were noted. Statistically significant differences were observed among control group and moderately differentiated adenocarcinomas and between all groups of adenocarcinomas. In two patients with sarcoma uteri the lowest EGF (0.49 and 0.58 ng/ml) serum concentrations were found. Our results confirm that low EGF serum concentrations as well as EGF receptor on the cell surface probably may increase the risk of cancerogenesis in human endometrium. Topics: Adenocarcinoma; Adult; Aged; Aged, 80 and over; Endometrial Neoplasms; Epidermal Growth Factor; Female; Humans; Middle Aged; Sarcoma | 1996 |
68 other study(ies) available for epidermal-growth-factor and Endometrial-Neoplasms
Article | Year |
---|---|
The ERK5/NF-κB signaling pathway targets endometrial cancer proliferation and survival.
Endometrial cancer (EC) is the most common type of gynecologic cancer in women of developed countries. Despite surgery combined with chemo-/radiotherapy regimens, overall survival of patients with high-risk EC tumors is poor, indicating a need for novel therapies. The MEK5-ERK5 pathway is activated in response to growth factors and to different stressors, including oxidative stress and cytokines. Previous evidence supports a role for the MEK5-ERK5 pathway in the pathology of several cancers. We investigated the role of ERK5 in EC. In silico analysis of the PanCancer Atlas dataset showed alterations in components of the MEK5-ERK5 pathway in 48% of EC patients. Here, we show that ERK5 inhibition or silencing decreased EGF-induced EC cell proliferation, and that genetic deletion of MEK5 resulted in EC impaired proliferation and reduced tumor growth capacity in nude mice. Pharmacologic inhibition or ERK5 silencing impaired NF-kB pathway in EC cells and xenografts. Furthermore, we found a positive correlation between ERK5 and p65/RELA protein levels in human EC tumor samples. Mechanistically, genetic or pharmacologic impairment of ERK5 resulted in downregulation of NEMO/IKKγ expression, leading to impaired p65/RELA activity and to apoptosis in EC cells and xenografts, which was rescued by NEMO/IKKγ overexpression. Notably, ERK5 inhibition, MEK5 deletion or NF-kB inhibition sensitized EC cells to standard EC chemotherapy (paclitaxel/carboplatin) toxicity, whereas ERK5 inhibition synergized with paclitaxel to reduce tumor xenograft growth in mice. Together, our results suggest that the ERK5-NEMO-NF-κB pathway mediates EC cell proliferation and survival. We propose the ERK5/NF-κB axis as new target for EC treatment. Topics: Animals; Carboplatin; Cell Proliferation; Cytokines; Endometrial Neoplasms; Epidermal Growth Factor; Female; Humans; MAP Kinase Kinase 5; MAP Kinase Signaling System; Mice; Mice, Nude; NF-kappa B; Paclitaxel | 2022 |
EpCAM-Regulated Transcription Exerts Influences on Nanomechanical Properties of Endometrial Cancer Cells That Promote Epithelial-to-Mesenchymal Transition.
Overexpression of epithelial cell adhesion molecule (EpCAM) has been implicated in advanced endometrial cancer, but its roles in this progression remain to be elucidated. In addition to its structural role in modulating cell-surface adhesion, here we demonstrate that EpCAM is a regulatory molecule in which its internalization into the nucleus turns on a transcription program. Activation of EGF/EGFR signal transduction triggered cell-surface cleavage of EpCAM, leading to nuclear internalization of its cytoplasmic domain EpICD. ChIP-seq analysis identified target genes that are coregulated by EpICD and its transcription partner, LEF-1. Network enrichment analysis further uncovered a group of 105 genes encoding functions for tight junction, adherent, and cell migration. Furthermore, nanomechanical analysis by atomic force microscopy revealed increased softness and decreased adhesiveness of EGF-stimulated cancer cells, implicating acquisition of an epithelial-mesenchymal transition (EMT) phenotype. Thus, genome editing of EpCAM could be associated with altering these nanomechanical properties towards a less aggressive phenotype. Using this integrative genomic-biophysical approach, we demonstrate for the first time an intricate relationship between EpCAM-regulated transcription and altered biophysical properties of cells that promote EMT in advanced endometrial cancer. Cancer Res; 76(21); 6171-82. ©2016 AACR. Topics: Biomechanical Phenomena; Cell Line, Tumor; Cell Nucleus; Endometrial Neoplasms; Epidermal Growth Factor; Epithelial Cell Adhesion Molecule; Epithelial-Mesenchymal Transition; Female; Gene Editing; Humans; Lymphoid Enhancer-Binding Factor 1; Microscopy, Atomic Force; Tight Junctions; Transcription, Genetic | 2016 |
An early-screening biomarker of endometrial carcinoma: NGAL is associated with epithelio-mesenchymal transition.
Neutrophil gelatinase-associated lipocalin is currently one of the most interesting and enigmatic proteins involved in the development of malignancies. In this study, we found that the expression of neutrophil gelatinase-associated lipocalin was up-regulated in endometrial cancer tissues and cell lines, significantly increased in early-grade ones, suggesting it may serve as a biomarker for early-stage screening for endometrial carcinoma. Moreover, neutrophil gelatinase-associated lipocalin was up-regulated in Ishikawa cells under going epithelio-mesenchymal transition induced by epidermal growth factor (5 ng/ml). Up-regulation of neutrophil gelatinase-associated lipocalin may correlate with the down-regulation of E-cadherin expression, up-regulation of Vimentin expression, enhanced cell migration, invasion and proliferation, which are the typical hallmarks of epithelio-mesenchymal transition processes. Neutrophil gelatinase-associated lipocalin may play a dual role during tumorigenetic and developmental processes of endometrial carcinoma. These results suggested neutrophil gelatinase-associated lipocalin to be a potential molecular target in the early diagnosis and treatment of endometrial carcinoma. Further studies are warranted to clarify the molecular mechanisms behind the expression and function of neutrophil gelatinase-associated lipocalin and epithelio-mesenchymal transition. Topics: Adult; Aged; Biomarkers, Tumor; Cell Movement; Early Detection of Cancer; Endometrial Neoplasms; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; Female; Humans; Lipocalin-2; Middle Aged; Neoplasm Invasiveness | 2016 |
A 78-year-old woman with brain metastases.
Topics: Adenocarcinoma; Aged; Anaplastic Lymphoma Kinase; Brain Neoplasms; Endometrial Neoplasms; Epidermal Growth Factor; Female; Genotype; Humans; Lung Neoplasms; Precision Medicine; Radiography; Receptor Protein-Tyrosine Kinases | 2015 |
GnRH-(1-5) activates matrix metallopeptidase-9 to release epidermal growth factor and promote cellular invasion.
In the extracellular space, the gonadotropin-releasing hormone (GnRH) is metabolized by the zinc metalloendopeptidase EC3.4.24.15 (EP24.15) to form the pentapeptide, GnRH-(1-5). GnRH-(1-5) diverges in function and mechanism of action from GnRH in the brain and periphery. GnRH-(1-5) acts on the orphan G protein-coupled receptor 101 (GPR101) to sequentially stimulate epidermal growth factor (EGF) release, phosphorylate the EGF receptor (EGFR), and facilitate cellular migration. These GnRH-(1-5) actions are dependent on matrix metallopeptidase (MMP) activity. Here, we demonstrated that these GnRH-(1-5) effects are dependent on increased MMP-9 enzymatic activity in the Ishikawa and ECC-1 cell lines. Furthermore, the effects of GnRH-(1-5) mediated by GPR101 and the subsequent increase in MMP-9 enzymatic activity lead to an increase in cellular invasion. These results suggest that GnRH-(1-5) and GPR101 regulation of MMP-9 may have physiological relevance in the metastatic potential of endometrial cancer cells. Topics: Cell Line, Tumor; Cell Movement; Endometrial Neoplasms; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; Gonadotropin-Releasing Hormone; Humans; Matrix Metalloproteinase 9; Neoplasm Metastasis; Peptide Fragments; Phosphorylation; Receptors, G-Protein-Coupled; Signal Transduction | 2015 |
GnRH-(1-5) transactivates EGFR in Ishikawa human endometrial cells via an orphan G protein-coupled receptor.
The decapeptide GnRH is known for its central role in the regulation of the hypothalamo-pituitary-gonadal axis. In addition, it is also known to have local effects within peripheral tissues. The zinc metalloendopeptidase, EC 3.4.24.15 (EP24.15), can cleave GnRH at the Tyr(5)-Gly(6) bond to form the pentapeptide, GnRH-(1-5). The central and peripheral effect of GnRH-(1-5) is different from its parent peptide, GnRH. In the current study, we examined the effect of GnRH-(1-5) on epidermal growth factor receptor (EGFR) phosphorylation and cellular migration. Using the Ishikawa cell line as a model of endometrial cancer, we demonstrate that GnRH-(1-5) stimulates epidermal growth factor release, increases the phosphorylation of EGFR (P < .05) at three tyrosine sites (992, 1045, 1068), and promotes cellular migration. In addition, we also demonstrate that these actions of GnRH-(1-5) are mediated by the orphan G protein-coupled receptor 101 (GPR101). Down-regulation of GPR101 expression blocked the GnRH-(1-5)-mediated release of epidermal growth factor and the subsequent phosphorylation of EGFR and cellular migration. These results suggest that GPR101 is a critical requirement for GnRH-(1-5) transactivation of EGFR in Ishikawa cells. Topics: Calcium Signaling; Cell Line, Tumor; Cell Movement; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression; Gonadotropin-Releasing Hormone; Humans; Matrix Metalloproteinase Inhibitors; Oligopeptides; Oncogene Proteins; Peptide Fragments; Phenylalanine; Phosphorylation; Protein Processing, Post-Translational; Pyrrolidonecarboxylic Acid; Quinazolines; Receptors, Cell Surface; Receptors, G-Protein-Coupled; Receptors, LHRH; Thiophenes; Transcriptional Activation; Tyrphostins | 2014 |
Mammalian sterile-like 1 kinase inhibits TGFβ and EGF‑dependent regulation of invasiveness, migration and proliferation of HEC-1-A endometrial cancer cells.
Transforming growth factor-β (TGFβ) and epidermal growth factor (EGF) are two potent regulators of tumorigenesis. Signaling cross-talk of TGFβ and EGF employs a number of regulators which define the impact on cell physiology. MST1 has recently been reported as a regulator of tumorigenesis and differentiation. To investigate the role of mammalian sterile-like 1 (MST1) in TGFβ and EGF signaling, we established transiently MST1‑transfected HEC-1-A endometrial cancer cells, and subjected the cells to treatment with TGFβ1, EGF and their combination. We report MST1 as a negative regulator of combined TGFβ and EGF signaling. We observed that enhanced expression of MST1 inhibited the combined action of TGFβ1 and EGF on cell invasiveness, migration and proliferation. Monitoring of the intracellular regulatory proteins showed that MST1 contribution to the TGFβ-EGF cross-talk may involve focal adhesion kinase and E-cadherin, but not activation of Smad2. Our data unveiled the role of MST1 as a negative feedback for TGFβ1‑ and EGF‑regulated cell invasiveness, migration and proliferation. Topics: Apoptosis; Cell Line, Tumor; Cell Movement; Cell Proliferation; Endometrial Neoplasms; Endometrium; Epidermal Growth Factor; Female; Hepatocyte Growth Factor; Humans; Immunoblotting; Neoplasm Invasiveness; Proto-Oncogene Proteins; Receptor Cross-Talk; Transfection; Transforming Growth Factor beta | 2014 |
[Expression of epidermal growth factor receptor and concentrations of epidermal growth factor and melatonin in endometrial carcinoma].
The aim of the research was to study the Epidermal Growth Factor Receptor (EGFR) expression, measure Epidermal Growth Factor (EGF) and melatonin concentrations in patients with endometrial carcinomas. In order to reveal EGFR expression, immunohistochemical examination of morphologic material (tumor of the uterine body, histopathological diagnosis: endometrial carcinoma) from 21 patients was performed. The EGF blood plasma levels were measured using the method of high performance liquid chromatography (HPLC).The blood serum levels of melatonin were measured by the method of immunoferment analysis ELISA(IBL International reagent). The resultant numerical data were processed statistically using the computer program SPSS-12-ANOVA. A statistical analysis was also performed using the system IBM SPSS Statistics, version 20. The investigation revealed: 1. In endometrial adenocarcinomas (high-grade, low-grade) the expression of EGFR is detected in 100%. 2. In endometrial adenocarcinomas a relatively weak positive correlation between EGFR expression and melatonin blood serum levels is observed. With that, there is a negative correlation between the blood levels of EGF and melatonin. 3. The average index of EGF blood plasma level in simple endometrial hyperplasia (atypia-free) is significantly lower than that of the patients with endometrial adenocarcinoma, while in complex endometrial hyperplasia (atypia-free) the average index of EGF blood plasma level is slightly lower than in endometrial carcinoma. With low-grade endometrial carcinomas, the average index of EGF blood plasma level is higher compared to that of the patients with high-grade endometrial adenocarcinomas. 4. The average level of blood plasma melatonin in patients with simple/complex endometrial hyperplasia (atypia-free) is significantly higher than in high-grade endometrial adenocarcinoma. 5. In endometrial adenocarcinoma (high-grade, low-grade),a drastic increase in average levels of blood plasma EGF is observed with a correspondingly sharp lowering in average blood serum melatonin levels. 6. Under about equal conditions (strong expression of EGFR in the timorous tissue and sharp/extremely sharp decrease in the levels of anti-proliferative/anti-neoplastic melatonin in blood serum) high values of proliferative EGF content in blood plasma are indicative of poor diagnosis. Topics: Adult; Aged; Endometrial Neoplasms; Enzyme-Linked Immunosorbent Assay; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression Regulation, Neoplastic; Humans; Melatonin; Middle Aged; Neoplasm Staging | 2014 |
Promoter hypomethylation of EpCAM-regulated bone morphogenetic protein gene family in recurrent endometrial cancer.
Epigenetic regulation by promoter methylation plays a key role in tumorigenesis. Our goal was to investigate whether altered DNA methylation signatures associated with oncogenic signaling delineate biomarkers predictive of endometrial cancer recurrence.. Methyl-CpG-capture sequencing was used for global screening of aberrant DNA methylation in our endometrial cancer cohort, followed by validation in an independent The Cancer Genome Atlas (TCGA) cohort. Bioinformatics as well as functional analyses in vitro, using RNA interference (RNAi) knockdown, were performed to examine regulatory mechanisms of candidate gene expression and contribution to aggressive phenotype, such as epithelial-mesenchymal transition (EMT).. We identified 2,302 hypermethylated loci in endometrial tumors compared with control samples. Bone morphogenetic protein (BMP) family genes, including BMP1, 2, 3, 4, and 7, were among the frequently hypermethylated loci. Interestingly, BMP2, 3, 4, and 7 were less methylated in primary tumors with subsequent recurrence and in patients with shorter disease-free interval compared with nonrecurrent tumors, which was validated and associated with poor survival in the TCGA cohort (BMP4, P = 0.009; BMP7, P = 0.007). Stimulation of endometrial cancer cells with epidermal growth factor (EGF) induced EMT and transcriptional activation of these genes, which was mediated by the epithelial cell adhesion molecule (EpCAM). EGF signaling was implicated in maintaining the promoters of candidate BMP genes in an active chromatin configuration and thus subject to transcriptional activation.. Hypomethylation signatures of candidate BMP genes associated with EpCAM-mediated expression present putative biomarkers predictive of poor survival in endometrial cancer. Topics: Antigens, Neoplasm; Base Sequence; Bone Morphogenetic Proteins; Cell Adhesion Molecules; Cell Line, Tumor; CpG Islands; Disease-Free Survival; DNA Methylation; Endometrial Neoplasms; Epidermal Growth Factor; Epithelial Cell Adhesion Molecule; Epithelial-Mesenchymal Transition; Female; Humans; Neoplasm Recurrence, Local; Promoter Regions, Genetic; RNA Interference; RNA, Small Interfering; Sequence Analysis, DNA; Survival; Transcriptional Activation | 2013 |
MUC20 overexpression predicts poor prognosis and enhances EGF-induced malignant phenotypes via activation of the EGFR-STAT3 pathway in endometrial cancer.
Mucins play a critical role in the malignancy of various tumors and have been identified as diagnostic markers and as attractive therapeutic targets. However, the role of mucin (MUC) 20 in endometrial cancer (EC) is still unknown.. The relationship between MUC20 expression and clinical characteristics of EC was analyzed in 97 EC tumors and 16 normal tissues by immunohistochemistry. Effects of MUC20 on EC cells, HEC-1A and RL95-2, were examined by in vitro cell growth, migration, and invasion assays, as well as in vivo tumor growth in SCID mouse model. Western blotting was performed to analyze signaling pathways modulated by MUC20.. MUC20 expression was significantly higher in EC tumors compared with the normal tissue. High levels of MUC20 expression in EC tumors were correlated with an unfavorable histologic subtype. Furthermore, MUC20 was an independent prognostic factor for poor survival as evaluated by multivariate analyses. Overexpression of MUC20 in EC cells significantly enhanced cell growth, migration, and invasion, as well as tumor growth in vivo. The MUC20-enhanced invasive behavior was significantly blocked by erlotinib, an EGFR inhibitor. Moreover, MUC20 overexpression enhanced EGF-mediated migration and invasion, suggesting a critical role of EGFR in MUC20-mediated effects. We found that MUC20 overexpression could enhance EGF-induced phosphorylation of EGFR and STAT3. Inhibition of the STAT3 activity by its inhibitor Stattic significantly suppressed the MUC20-enhanced invasive behavior.. MUC20 is novel prognostic factor for EC and its overexpression enhances EGF-triggered invasive behavior through activation of EGFR-STAT3 pathway. Topics: Animals; Cell Growth Processes; Cell Line, Tumor; Disease Models, Animal; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Mice; Mice, SCID; Middle Aged; Mucins; Neoplasm Staging; Phenotype; Phosphorylation; Prognosis; Signal Transduction; STAT3 Transcription Factor; Transfection | 2013 |
Serum levels of epidermal growth factor, transforming growth factor, and c-erbB2 in ovarian cancer.
This study aimed to investigate serum levels of epidermal growth factor (EGF), transforming growth factor α (TGF-α), and c-erbB2 in patients with ovarian cancer.. In this retrospective cohort study, the study and control groups were composed of 43 women with a prediagnosis of ovarian cancer and 43 healthy women, respectively. Blood samples from all women were obtained and studied by enzyme-linked immunosorbent assay kits for EGF, TGF-α, and c-erbB2. After surgery of the study group, ovarian cancer was confirmed and compared with control group. Stage, grade, and histological types were defined after histopathologic examination, and subgroups were constructed and compared.. Serum EGF, TGF-α, and c-erbB2 levels were significantly increased in study group compared with those in the control group (P < 0.001). There were no differences in serum levels of EGF, TGF-α, and c-erbB2 among all stages, grades, and histological types of ovarian cancer. If 47.90 pg/mL was selected as the cutoff value, EGF has an 80% sensitivity and a 65% specificity for detecting ovarian cancer. The cutoff value of 41,095.00 pg/mL for TGF-α has a 90% sensitivity and a 72% specificity for detecting ovarian cancer. The c-erbB2 level of 4.63 pg/mL as the cutoff value has an 83% sensitivity and a 76% specificity for predicting ovarian cancer.. Serum levels of EGF, TGF-α, and c-erbB2 may be used for diagnosing ovarian cancer. Topics: Adenocarcinoma, Clear Cell; Adenocarcinoma, Mucinous; Biomarkers, Tumor; Case-Control Studies; Cystadenocarcinoma, Serous; Endometrial Neoplasms; Enzyme-Linked Immunosorbent Assay; Epidermal Growth Factor; Female; Humans; Middle Aged; Neoplasm Grading; Neoplasm Staging; Ovarian Neoplasms; Prognosis; Receptor, ErbB-2; Retrospective Studies; ROC Curve; Transforming Growth Factor alpha | 2012 |
High-risk endometrial carcinoma profiling identifies TGF-β1 as a key factor in the initiation of tumor invasion.
Endometrial cancer is among the three most common cancers in females in industrialized countries. In the majority of cases, the tumor is confined to the uterus at the time of diagnosis and presents a good prognosis. However, after primary surgery, 15% to 20% of these tumors recur and have limited response to systemic therapy. We carried out gene expression profiling of high-risk recurrence endometrial cancers to identify new therapeutic approaches targeting the molecular pathways involved in the acquisition of an aggressive tumor phenotype. A microarray gene-expression analysis on a total of 51 human endometrial carcinomas revealed 77 genes specifically altered in high-risk recurrence tumors (P < 0.001). The bioinformatics analysis of gene-gene interactions and molecular relationships among these genes pointed to a prominent role for TGF-β1 signaling in the acquisition of an aggressive phenotype. We further showed that TGF-β1 has a principal role at the initiation of endometrial carcinoma invasion through the promotion of the epithelial to mesenchymal transition that leads to the acquisition of an invasive phenotype in HEC-1A and RL95-2 cells. Impairment of this initial step with SB-431542, a specific TGF-β1 inhibitor, precluded further persistent endometrial carcinoma invasion. In conclusion, we showed that the characterization of the molecular changes associated with the acquisition of an aggressive phenotype represents a realistic strategy for the rational identification and characterization of new potential therapeutic targets in an effort to improve the clinical management and the outcome of high-risk endometrial cancer patients. Topics: Benzamides; Cell Line, Tumor; Dioxoles; Endometrial Neoplasms; Epidermal Growth Factor; Epithelial-Mesenchymal Transition; Female; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; Models, Biological; Neoplasm Invasiveness; Recurrence; Signal Transduction; Transforming Growth Factor beta1 | 2011 |
ER-α36, a novel variant of estrogen receptor α, is involved in EGFR-related carcinogenesis in endometrial cancer.
To explore the role of estrogen receptor-α36 (ER-α36) in epidermal growth factor receptor (EGFR)-related carcinogenesis in endometrial cancer.. The expression of ER-α36, EGFR, and phospho-extracellular signal-regulated kinase was analyzed using immunohistochemistry in endometrial cancer samples. The cellular localization of ER-α36 and EGFR was determined using immunofluorescence in the endometrial cancer Hec1A cells. The level of phospho-extracellular signal-regulated kinase of Hec1A cells was determined using Western blotting after treatment with epidermal growth factor.. Positive rate of ER-α36 was increased in high-stage (P = .03) and high-grade (P = .224) endometrial cancer; expression of ER-α36 and EGFR exhibited a significant positive correlation (r = 0.334, P = .025) and they showed substantial colocalization on the plasma membrane of glandular cells; phospho-extracellular signal-regulated kinase positive rate in ER-α36 positive group and EGFR positive group was higher than that of ER-α36 negative group (P = .014) and EGFR negative group (P = .016); finally, ER-α36 mediated epidermal growth factor-stimulated extracellular signal-regulated kinase activation in Hec1A cells.. ER-α36 mediates EGFR-related extracellular signal-regulated kinase activation in endometrial cancer. Topics: Cell Line, Tumor; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Estrogen Receptor alpha; Extracellular Signal-Regulated MAP Kinases; Female; Humans; Phosphorylation; Protein Isoforms; Signal Transduction | 2011 |
SLC37A1 gene expression is up-regulated by epidermal growth factor in breast cancer cells.
Phospholipid biosynthesis exerts an important role in the proliferation of tumor cells; however, the regulation of the proteins involved in this context still remains to be fully evaluated. SLC37A1 protein belongs to a small family of sugar-phosphate/phosphate exchangers. The sequence homology with the bacterial glycerol-3-phosphate transporter (30%) suggests that SLC37A1 might be able to catalyze an exchange of glycerol-3-phosphate against phosphate. Glycerol-3-phosphate, found in different cellular compartments, is a fundamental substrate in phospholipid biosynthesis. In the present study, we demonstrate for the first time that epidermal growth factor (EGF) transactivates SLC37A1 promoter sequence and induces SLC37A1 mRNA, and protein expression through the EGFR/MAPK/Fos transduction pathway in ER-negative SkBr3 breast cancer cells. These findings were corroborated by comparable results obtained in ER-positive endometrial Ishikawa tumor cells. Interestingly, we also show that SLC37A1 protein localizes in the endoplasmic reticulum, hence supporting its possible involvement in phospholipid biosynthesis. On the basis of our data, the up-regulation of SLC37A1 gene expression should be included among the well-known stimulatory action exerted by EGF in breast cancer cells. In addition, further studies are required to provide evidence concerning the potential role of EGF-mediated SLC37A1 induction in breast tumor cells. Topics: Base Sequence; Blotting, Western; Breast Neoplasms; Chromatin Immunoprecipitation; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Fluorescent Antibody Technique; Humans; Immunoenzyme Techniques; Luciferases; Male; Membrane Transport Proteins; Molecular Sequence Data; Promoter Regions, Genetic; Prostatic Neoplasms; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Tumor Cells, Cultured; Up-Regulation | 2010 |
Olfactomedin-4 regulation by estrogen in the human endometrium requires epidermal growth factor signaling.
Olfactomedin-4 (OLFM-4) is an extracellular matrix protein that is highly expressed in human endometrium. We have examined the regulation and function of OLFM-4 in normal endometrium and in cases of endometriosis and endometrial cancer. OLFM-4 expression levels are highest in proliferative-phase endometrium, and 17β-estradiol up-regulates OLFM-4 mRNA in endometrial explant cultures. Using the luciferase reporter under control of the OLFM-4 promoter, it was shown that both 17β-estradiol and OH-tamoxifen induce luciferase activity, and epidermal growth factor receptor-1 is required for this estrogenic response. In turn, EGF activates the OLFM-4 promoter, and estrogen receptor-α is needed for the complete EGF response. The cellular functions of OLFM-4 were examined by its expression in OLFM-4-negative HEK-293 cells, which resulted in decreased vimentin expression and cell adherence as well as increased apoptosis resistance. In cases of endometriosis and endometrial cancer, OLFM-4 expression correlated with the presence of epidermal growth factor receptor-1 and estrogen receptor-α (or estrogen signaling). An increase of OLFM-4 mRNA was observed in the endometrium of endometriosis patients. No change in OLFM-4 expression levels were observed in patients with endometrial cancer relative with controls. In conclusion, cross-talk between estrogen and EGF signaling regulates OLFM-4 expression. The role of OLFM-4 in endometrial tissue remodeling before the secretory phase and during the predisposition and early events in endometriosis can be postulated but requires additional investigation. Topics: Adult; Aged; Aged, 80 and over; Apoptosis; Cell Adhesion; Cells, Cultured; Endometrial Neoplasms; Endometriosis; Endometrium; Epidermal Growth Factor; ErbB Receptors; Estrogens; Female; Granulocyte Colony-Stimulating Factor; Humans; Menstrual Cycle; Middle Aged; Promoter Regions, Genetic; Signal Transduction; Trefoil Factor-1; Tumor Suppressor Proteins; Vimentin | 2010 |
Early effects of high concentrations of progesterone and mifepristone A gene expression study of endometrial cancer cells (Ishikawa).
Patients with endometrial hyperplasia representing preliminary stages of endometrial cancer have shown to respond to therapy in 100% of the cases when treated with levonorgestrel-impregnated intrauterine device. Anti-proliferative effect has also been reported after application of an anti-progestin impregnated intrauterine device which showed to induce endometrial atrophy. The intention of the present study was to obtain more information of novel therapeutic targets for hormonal treatment in endometrial hyperplasia and endometrial cancers. Gene expression of signaling pathways after stimulation of Ishikawa cells with high doses of progesterone (32 microM) or Mifepristone (32 microM) was performed. After using an oligo microarrays representing 24,650 human genes and 37,580 gene transcripts, 6154 genes remained after pre-processing and filtering. This resulted in a total of 993 up-regulated genes with 189 genes for progesterone and 255 genes for Mifepristone. The 550 down-regulated genes were distributed with 256 genes for progesterone, 127 genes for RU 486. The results showed that genes presenting the epidermal growth factor (EGF)/MAP-kinase pathway were significantly over-represented by progesterone treatment, whereas, by Mifepristone treatment genes involved in the p53 pathway were also up-regulated (data not shown). These genes may be interesting as potential new therapeutic targets in endometrial hyperplasia and endometrial cancer, as candidate genes for therapy response or as candidate markers for tumor progression. Topics: Cell Line, Tumor; Down-Regulation; Endometrial Neoplasms; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; Humans; Mifepristone; Progesterone; Progestins; Receptors, Progesterone; Up-Regulation | 2009 |
Common genetic variability in ESR1 and EGF in relation to endometrial cancer risk and survival.
We investigated common genetic variation in the entire ESR1 and EGF genes in relation to endometrial cancer risk, myometrial invasion and endometrial cancer survival. We genotyped a dense set of single-nucleotide polymorphisms (SNPs) in both genes and selected haplotype tagging SNPs (tagSNPs). The tagSNPs were genotyped in 713 Swedish endometrial cancer cases and 1567 population controls and the results incorporated into logistic regression and Cox proportional hazards models. We found five adjacent tagSNPs covering a region of 15 kb at the 5' end of ESR1 that decreased the endometrial cancer risk. The ESR1 variants did not, however, seem to affect myometrial invasion or endometrial cancer survival. For the EGF gene, no association emerged between common genetic variants and endometrial cancer risk or myometrial invasion, but we found a five-tagSNP region that covered 51 kb at the 5' end of the gene where all five tagSNPs seemed to decrease the risk of dying from endometrial cancer. One of the five tagSNPs in this region was in strong linkage disequilibrium (LD) with the untranslated A61G (rs4444903) EGF variant, earlier shown to be associated with risk for other forms of cancer. Topics: Aged; Case-Control Studies; Endometrial Neoplasms; Epidermal Growth Factor; Estrogen Receptor alpha; Female; Genetic Variation; Genotype; Humans; Middle Aged; Neoplasm Invasiveness; Polymorphism, Single Nucleotide; Registries; Risk Factors; Survival Analysis; Sweden | 2009 |
Activation of epidermal growth factor receptor results in snail protein but not mRNA overexpression in endometrial cancer.
Reduced E-cadherin expression is associated with tumour progression of many carcinomas, including endometrial cancers. The transcription factor Snail is known as one of the most prominent transcriptional E-cadherin repressors; its regulation in cancer tissues, however, still remains unclear. Here, we report that activation of epidermal growth factor receptor (EGFR) resulted in overexpression of Snail and also identified critical downstream signalling molecules. Stimulation of two endometrial carcinoma cell lines with epidermal growth factor (EGF) lead to an increase of Snail protein expression. In primary human endometrioid endometrial carcinomas Snail protein expression correlated with the activated, phosphorylated form of EGFR (Tyr1086) as revealed by profiling 24 different signalling proteins using protein lysate microarrays. In addition, we observed an inverse correlation between Snail and E-cadherin protein levels in these tumours. Most likely, p38 MAPK, PAK1, AKT, ERK1/2 and GSK-3beta are involved in the up-regulation of Snail downstream of EGFR. Snail mRNA expression did not show a correlation with activated EGFR in these tumours. Taken together, profiling of signalling proteins in primary human tissues provided strong evidence that EGFR signalling is involved in Snail protein overexpression. Topics: Blotting, Western; Cell Line, Tumor; Cluster Analysis; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression Regulation, Neoplastic; Humans; Oligonucleotide Array Sequence Analysis; Phosphorylation; Pilot Projects; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Snail Family Transcription Factors; Transcription Factors | 2009 |
Expression of nodal signalling components in cycling human endometrium and in endometrial cancer.
The human endometrium is unique in its capacity to remodel constantly throughout adult reproductive life. Although the processes of tissue damage and breakdown in the endometrium have been well studied, little is known of how endometrial regeneration is achieved after menstruation. Nodal, a member of the transforming growth factor-beta superfamily, regulates the processes of pattern formation and differentiation that occur during early embryo development.. In this study, the expression of Nodal, Cripto (co-receptor) and Lefty A (antagonist) was examined by RT-PCR and immunohistochemistry across the menstrual cycle and in endometrial carcinomas.. Nodal and Cripto were found to be expressed at high levels in both stromal and epithelial cells during the proliferative phase of the menstrual cycle. Although immunoreactivity for both proteins in surface and glandular epithelium was maintained at relatively steady-state levels across the cycle, their expression was significantly decreased within the stromal compartment by the mid-secretory phase. Lefty expression, as has previously been reported, was primarily restricted to glandular epithelium and surrounding stroma during the late secretory and menstrual phases. In line with recent studies that have shown that Nodal pathway activity is upregulated in many human cancers, we found that Nodal and Cripto immunoreactivity increased dramatically in the transition from histologic Grade 1 to histologic Grades 2 and 3 endometrial carcinomas. Strikingly, Lefty expression was low or absent in all cancer tissues.. The expression of Nodal in normal and malignant endometrial cells that lack Lefty strongly supports an important role for this embryonic morphogen in the tissue remodelling events that occur across the menstrual cycle and in tumourogenesis. Topics: Adult; Body Fluids; Carcinoma; Endometrial Neoplasms; Endometrium; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; GPI-Linked Proteins; Humans; Intercellular Signaling Peptides and Proteins; Left-Right Determination Factors; Membrane Glycoproteins; Menstrual Cycle; Neoplasm Proteins; Nodal Protein; Signal Transduction; Uterus | 2009 |
FGF2 transcript levels are positively correlated with EGF and IGF-1 in the malignant endometrium.
Epidermal and insulin-like growth factors (EGF, IGFs) act as mitogens promoting endothelial cell proliferation and differentiation. Accumulating evidence for interactions between EGF and IGF signaling pathways has been reported. Fibroblast growth factor-2 (FGF2) is also mitogenic for various cell types and is associated with regulation of tumor angiogenesis and metastasis. However EGF, IGF-1 and FGF2 transcript levels have been scarcely investigated in endometrial carcinoma.. In the present study, we evaluated the mRNA expression pattern of EGF, IGF-1 and FGF2 by using Comparative Quantitative real-time RT-PCR assay in 30 endometrial cancer specimens and an equal number of adjacent normal tissues.. Both overexpression and down-regulation of EGF, IGF-1 and FGF2 was demonstrated in endometrial cancer compared to the adjacent normal specimens; however the main features of cancer were IGF-1 and EGF down-regulation and FGF2 up-regulation. Different co-expression patterns of all three factors were displayed in normal and malignant endometrium. Interestingly, FGF2 mRNA was positively correlated with EGF and IGF-1 transcript levels in endometrial cancer (P=0.024 and P=0.006, respectively), while no co-expression was observed in the adjacent normal specimens. Furthermore, endometrial tissue-pair analysis revealed a significant positive correlation between EGF and IGF-1 (P=0.010), supporting the hypothesis of a cross-talk between IGF- and EGF-mediated signaling pathways in endometrial cancer. EGF transcript levels were marginally higher in endometrioid than non-endometrioid tumors (P=0.050), and in grade I compared to grade II tumors (P=0.053).. Up-regulation as well as down-regulation of EGF, IGF-1 and FGF2 transcript levels is observed in endometrial cancer; however IGF-1 and EGF down-regulation and FGF2 up-regulation seem to comprise the main features of endometrial carcinogenesis. The disruption of their mRNA co-expression pattern observed supports the hypothesis of a cross-talk between IGF- and EGF-mediated signaling pathways in promoting endothelial cell proliferation and differentiation in endometrial cancer. Topics: Adult; Aged; Aged, 80 and over; Carcinoma, Endometrioid; Down-Regulation; Endometrial Neoplasms; Endometrium; Epidermal Growth Factor; Female; Fibroblast Growth Factor 2; Gene Expression Regulation, Neoplastic; Humans; Insulin-Like Growth Factor I; Middle Aged; Polymerase Chain Reaction; RNA, Messenger; Up-Regulation | 2008 |
Epidermal-growth-factor-dependent phosphorylation and ubiquitinylation of MAGE-11 regulates its interaction with the androgen receptor.
The androgen receptor (AR) is a ligand-activated transcription factor that interacts with coregulatory proteins during androgen-dependent gene regulation. Melanoma antigen gene protein 11 (MAGE-11) is an AR coregulator that specifically binds the AR NH(2)-terminal FXXLF motif and modulates the AR NH(2)- and carboxyl-terminal N/C interaction to increase AR transcriptional activity. Here we demonstrate that epidermal growth factor (EGF) signaling increases androgen-dependent AR transcriptional activity through the posttranslational modification of MAGE-11. EGF in the presence of dihydrotestosterone stabilizes the AR-MAGE complex through the site-specific phosphorylation of MAGE-11 at Thr-360 and ubiquitinylation at Lys-240 and Lys-245. The time-dependent EGF-induced increase in AR transcriptional activity by MAGE-11 is mediated through AR activation functions 1 and 2 in association with the increased turnover of AR and MAGE-11. The results reveal a dynamic mechanism whereby growth factor signaling increases AR transcriptional activity through the covalent modification of an AR-specific coregulatory protein. Sequence conservation of the MAGE-11 phosphorylation and ubiquitinylation sites throughout the MAGE gene family suggests common regulatory mechanisms for this group of cancer-testis antigens. Topics: Adenocarcinoma; Amino Acid Motifs; Animals; Antigens, Neoplasm; Cell Line; Chlorocebus aethiops; COS Cells; Dihydrotestosterone; Endometrial Neoplasms; Epidermal Growth Factor; Female; HeLa Cells; Humans; Multiprotein Complexes; Neoplasm Proteins; Phosphorylation; Protein Interaction Mapping; Protein Processing, Post-Translational; Receptors, Androgen; Recombinant Fusion Proteins; Transcription, Genetic; Transfection; Ubiquitination | 2008 |
Expression of the epidermal growth factor system in endometrioid endometrial cancer.
The Epidermal Growth Factor (EGF) system is expressed in healthy premenopausal endometrium. We describe the expression of the four receptors, HER1, HER2, HER3, HER4 and the six ligands amphiregulin, transforming growth factor alpha (TGF-alpha), heparin binding EGF like growth factor (HB-EGF), betacellulin, epiregulin and EGF in endometrioid endometrial cancer.. We have uterine samples from 45 women with endometrioid endometrial cancer. As normal counterparts, endometrial samples from thirteen postmenopausal women, and previous data on fourteen premenopausal women were employed. Extracted RNA was analyzed by real-time PCR; the receptors and ligands were localized by immunohistochemistry.. Three receptors (HER1, HER2 and HER4) and two detectable ligands (TGF-alpha and HB-EGF) are expressed significantly higher in endometrial cancer than in healthy postmenopausal endometrium. Cancer tissue show significantly lower expression of HER1 and HER3, and higher expression of HER4, amphiregulin, TGF-alpha and HB-EGF compared to premenopausal endometrium; no difference is seen in HER2. EGF is undetectable in all of the samples. Immunohistochemically the receptors locate to the epithelium and/or glands while the ligands locate to the stroma (amphiregulin), the stroma and the epithelium (TGF-alpha, epiregulin), the epithelium (betacellulin) or are not detectable (HB-EGF, EGF).. mRNA of all receptors and five ligands are present in endometrioid endometrial cancer, and the protein of all receptors and four ligands are identified by immunohistochemistry. The expression pattern in endometrioid endometrial cancer differs from the pattern in pre- and postmenopausal endometrium. The most remarkable finding is an increased level of HER4, a receptor which correlates to a favorable prognosis in other types of cancers. Topics: Adult; Aged; Aged, 80 and over; Carcinoma, Endometrioid; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Immunohistochemistry; Middle Aged; RNA, Messenger | 2007 |
Activation of ERK1/2 occurs independently of KRAS or BRAF status in endometrial cancer and is associated with favorable prognosis.
The extracellular-regulated kinase (ERK) signaling pathway plays important roles in regulating the malignant potential of cancer cells in vitro. However, the effect of ERK signaling on the prognosis of human tumors is not clearly understood. The present study examined the expression of phosphorylated ERK1/2 (p-ERK1/2) as a hallmark of ERK activation, in relation to KRAS and BRAF mutations, in 63 endometrial cancer specimens with endometrioid-subtype, in order to clarify the prognostic value of p-ERK1/2 expression. Immmunohistochemical analysis revealed that 40 tumors (63%) expressed p-ERK1/2, with varying levels of expression. Total ERK1/2 expression was also evaluated in a subset of tumors; most cases expressed ERK1/2 constitutively but no correlation was observed with p-ERK expression, indicating that p-ERK1/2 staining was not due to ERK overexpression but to hyperactivation of ERK1/2. There was no statistically significant correlation between p-ERK1/2 expression and clinicopathological features, including patient age, International Federation of Gynecology and Obstetrics stage, pathological grade, myometrial invasion and lymph node metastasis. Sequencing analysis indicated that 23% of patients had a mutation in exon 1 of KRAS, whereas none of the patients had a mutation in exons 11 or 15 of BRAF, which are reportedly hot spots for mutation in many tumor types. There was no significant correlation between KRAS or BRAF status and p-ERK1/2 expression. Unexpectedly, patients with low p-ERK1/2 expression had significantly lower relapse-free survival (P = 0.041) and overall survival (P = 0.020). Multivariate Cox regression analysis indicated that p-ERK1/2 expression was an independent prognostic indicator for overall survival (P = 0.047). These findings suggest that ERK activation occurs in a KRAS- and BRAF-independent manner in endometrial cancer, and is associated with favorable prognosis. Topics: Adult; Aged; Blotting, Western; Butadienes; Cell Line; Endometrial Neoplasms; Enzyme Activation; Enzyme Inhibitors; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; Genes, ras; Humans; Immunohistochemistry; Kaplan-Meier Estimate; Middle Aged; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mutation; Nitriles; Phosphorylation; Prognosis; Proportional Hazards Models; Proto-Oncogene Proteins B-raf; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger | 2007 |
PTEN sensitizes epidermal growth factor-mediated proliferation in endometrial carcinoma cells.
The fact that the genetic alterations of PTEN are frequently found in hormone-dependent cancers, such as endometrial, breast, and prostate cancers, might suggest the involvement of PTEN in the hormone-dependent cell growth of such tumors. Estrogen promotes the cell growth of the tumors by inducing peptide growth factors in part. We analyzed the possible involvement of PTEN in peptide-growth factor-dependent cell growth in endometrial carcinoma cells. PTEN-null Ishikawa cells were efficiently infected with recombinant adenovirus at 20 MOI (multiplicity of infection) to express PTEN protein. In PTEN-IK cells, phospho-Akt/PKB was down-regulated regardless of the consistent expression of Akt/PKB. The cell growth of parental IK cells was significantly stimulated by EGF and IGF-I, and PTEN-IK cells were further sensitized to the EGF-or IGF-I-growth stimulation. EGFR antibody could completely compromise the stimulatory effects of EGF in both cell lines. Wortmannin, a PI3K inhibitor, or UO126, a MAPK inhibitor, partly suppressed EGF-mediated cell growth stimulation in both cell lines. EGF augmented the level of phospho-Akt/PKB of PTEN-IK cells more effectively than that of parental IK cells. These results imply that the dysfunction of PTEN leads cells into a less-sensitive phenotype to peptide growth factors by constitutive activation of the PI3K/Akt/PKB signaling pathway in endometrial carcinoma. Topics: Adenoviridae; Androstadienes; Antibodies; Blotting, Western; Butadienes; Cell Line; Cell Line, Tumor; Cell Proliferation; Cell Survival; DNA, Recombinant; Endometrial Neoplasms; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Female; Fibroblast Growth Factor 2; Gene Expression; Hepatocyte Growth Factor; Humans; Insulin-Like Growth Factor I; Mitogen-Activated Protein Kinases; Nitriles; Phosphoinositide-3 Kinase Inhibitors; Phosphorylation; Proto-Oncogene Proteins c-akt; PTEN Phosphohydrolase; Receptor, ErbB-2; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Transfection; Wortmannin | 2006 |
Effects of insulin-like growth factor (IGF) binding protein-3 (IGFBP-3) on endometrial cancer (HHUA) cell apoptosis and EGF stimulated cell proliferation in vitro.
IGFBP-3 has been demonstrated to stimulate or inhibit cell proliferation independently of its ability to bind IGF and a specific IGFBP-3 receptor has been proposed. EGF has been implicated in the cancer development and carcinogenesis. Only limited data are available on the crosstalk between IGFBP-3 signaling and EGF induced cell survival and signal transduction. The current studies were undertaken to characterize IGFBP-3 binding to endometrial cancer cells (HHUA) and determine its biological effects, as well as whether IGFBP-3 exposure alters the cell proliferation stimulated by EGF.. Cell proliferation and apoptosis were analyzed by ELISA using specific antibodies. The interaction between HHUA cell and IGFBP-3 was analyzed using a biosensor. The phosphorylation abundance of specific proteins and their phosphorylation in response to EGF and IGFBP-3 was analyzed by immunoprecipitation followed by immunoblotting.. Biosensor analysis showed that IGFBP-3 could bind to HHUA cell surface. IGFBP-3 inhibited BrdU uptake, potentiated ssDNA production and induced p53 in HHUA cells. Although EGF stimulated HHUA cell proliferation and Akt phosphorylation, IGFBP-3 inhibited cell proliferation and Akt phosphorylation that had been stimulated by EGF. However, EGF receptor phosphorylation and expression were not reduced by IGFBP-3. Since HHUA cells lack IGF receptors and do not show biological response to IGF these results suggest that IGFBP-3 can bind to HHUA cells, inhibit cell proliferation and induce apoptosis independently of its ability to bind to IGFs possibly by binding to an IGFBP-3 receptor.. Taken together these findings demonstrate that IGFBP-3 binds to HHUA cell surface, and inhibits cell division induced by EGF, possibly by modulating the EGF-mediated signal transduction system. Topics: Antimetabolites, Antineoplastic; Apoptosis; Biosensing Techniques; Bromodeoxyuridine; Cell Membrane; Cell Proliferation; DNA, Single-Stranded; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Insulin-Like Growth Factor Binding Protein 3; Phosphorylation; Proto-Oncogene Proteins c-akt | 2006 |
GnRH-II agonist [D-Lys6]GnRH-II inhibits the EGF-induced mitogenic signal transduction in human endometrial and ovarian cancer cells.
The majority of human endometrial and ovarian cancers express receptors for GnRH type I (GnRH-I). Their proliferation is time- and dose-dependently reduced by GnRH-I and its analogs. GnRH-I analogs activate a phosphotyrosine-phosphatase (PTP) and inhibit EGF-induced mitogenic signal transduction. Recently we found that GnRH type II (GnRH-II) and its agonist [D-Lys6]GnRH-II also have antiproliferative effects on these tumor cells which are significantly greater than those of GnRH-I agonists. In a more recent study, we showed that the antiproliferative activity of GnRH-II on human endometrial and ovarian cancer cell lines is not mediated through the GnRH-I receptor. The underlying signal transduction mechanisms of GnRH-II are still unknown. In this study we showed that the mitogenic effects of growth factors in endometrial and ovarian cancer cell lines were counteracted by GnRH-II agonist [D-Lys6]GnRH-II, indicating an interaction with the mitogenic signal transduction. We showed that [D-Lys6]GnRH-II reduces EGF-induced auto-tyrosine-phosphorylation of EGF-receptors via activation of a PTP and that EGF-induced activation of mitogen-activated protein kinase was blocked in cells treated with [D-Lys6]GnRH-II. Furthermore, EGF-induced expression of the immediate early gene c-fos was inhibited by treatment with [D-Lys6]GnRH-II. After knock-out of GnRH-I receptor expression, GnRH-II agonist [D-Lys6]GnRH-II still activated PTP and inhibited the EGF-induced mitogenic signal transduction. These data indicate, that the effects of GnRH-II are not due to a cross-reaction with the GnRH-I receptor. In conclusion these data suggest that the signaling of GnRH-II agonist [D-Lys6]GnRH-II is comparable to that of GnRH-I analogs. Topics: Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Humans; MAP Kinase Kinase Kinases; Ovarian Neoplasms; Protein Tyrosine Phosphatases; Proto-Oncogene Proteins c-fos; Receptors, LHRH; RNA, Messenger; Signal Transduction; Triptorelin Pamoate | 2006 |
Expression of the epidermal growth factor system in endometrial cancer after adjuvant tamoxifen treatment for breast cancer.
The aim of our study was to describe the expression of cerbB-1, cerbB-2, cerbB-3 and cerbB-4 in endometrial cancer tissue and its correlation with clinicopathologic features and prognosis of endometrial cancer patients diagnosed during or after tamoxifen treatment for breast cancer. Thirteen tamoxifen-related endometrial cancers were identified from the archives of the Department of Obstetrics and Gynecology of the University of Patras, Medical School. Tissue specimens from endometrial lesions were immunostained for cerbB-1, cerbB-2, cerbB-3 and cerbB-4. For cerbB-1, five cases were positive and eight were negative. For cerbB-2, ten cases were positive and three were negative. For cerbB-3, nine cases were positive and four were negative. For cerbB-4, eight cases were positive and five were negative. However, a limitation of our study is that the number of cases was small, and further investigations are necessary to allow a more focused evaluation of cerbB-1, cerbB-2, cerbB-3 and cerbB-4 status, as a prognostic factor for endometrial cancer after tamoxifen treatment. Topics: Aged; Aged, 80 and over; Antineoplastic Agents, Hormonal; Breast Neoplasms; Carcinoma, Endometrioid; Chemotherapy, Adjuvant; Endometrial Neoplasms; Epidermal Growth Factor; Female; Humans; Middle Aged; Neoplasm Staging; Postmenopause; Tamoxifen | 2006 |
The estrogen receptor (ER)alpha variant Delta5 exhibits dominant positive activity on ER-regulated promoters in endometrial carcinoma cells.
Estrogen receptor (ER)alpha is a ligand-inducible transcription factor that mediates the physiological effects of 17beta-estradiol (E2). In the uterus, E2 is involved in tissue growth, maintenance, and differentiation. Delta5ERalpha (Delta5) is an ERalpha variant protein expressed in uterine tumors but not in normal tissue. We examined the transcriptional activity of Delta5 and its modulation of human ERalpha basal and E2-stimulated activity in Ishikawa cells, an endometrial cancer cell line. In transient transfection assays, Delta5 increased basal activity of an estrogen response element-containing promoter in the absence or presence of ERalpha but lessened stimulation by ERalpha and E2. Effects of Delta5 were not limited to model reporters, given that cyclin D1 and complement 3 promoters were similarly affected. Increases in basal transcription required dimerization and DNA binding of Delta5, whereas decreased E2 stimulation with ERalpha required only DNA binding. Decreased ligand stimulation was not unique to E2 but also applied to the selective ER modulators tamoxifen and genistein. However, promoter stimulation by epidermal growth factor is retained with Delta5. The ERalpha coactivator small nuclear ring finger protein is expressed in Ishikawa cells and uterine tumors, and it enhances effects of Delta5 alone and with ERalpha on basal activity of an estrogen response element reporter. Thus, in the presence of Delta5 plus ERalpha, there is a lower transcriptional response to E2 and SERMS, but stimulation by epidermal growth factor is retained. The expression of Delta5 in uterine carcinoma may provide a mechanism by which tumors could maintain expression of E2-responsive genes in the absence of E2. Topics: Cell Line, Tumor; Dimerization; Endometrial Neoplasms; Epidermal Growth Factor; Estradiol; Estrogen Receptor alpha; Female; Gene Expression Regulation, Neoplastic; Humans; Ligands; Mutagenesis, Site-Directed; Nuclear Proteins; Promoter Regions, Genetic; Selective Estrogen Receptor Modulators; Serine; Uterus | 2005 |
Progesterone increases tissue factor gene expression, procoagulant activity, and invasion in the breast cancer cell line ZR-75-1.
Progesterone in hormonal preparations increases the incidence of breast cancer. Tissue factor (TF), the initiator of the extrinsic coagulation pathway, is associated with metastasis in a wide variety of cancers. We demonstrate herein that TF mRNA and protein are up-regulated by progesterone in the breast cancer cell line ZR-75. Epidermal growth factor, also associated with increased breast cancer risk, did not regulate TF. The increase in TF is both rapid and transient; increasing after 6 h, reaching a maximum at 24 h, before decreasing to basal levels at 72 h. Sucrose gradient experiments demonstrated that TF is located in the heavy fraction of the plasma membrane, although caveolin-1 is not expressed in ZR-75. To understand the physiological implications of an increase in TF, we performed coagulation and invasion assays. An increase in TF corresponded to an increase in procoagulant activity. Furthermore, progesterone increased the invasion of ZR-75 cells through a matrigel, an effect that was blocked by an antibody against TF. Because TF expression is associated with an enhanced risk of metastasis, we postulate that the progesterone-dependent up-regulation of TF provides a survival advantage to burgeoning breast cancer cells and may contribute to the increased risk of cancer associated with combined hormone replacement therapy. Topics: Breast Neoplasms; Cell Line, Tumor; Cell Survival; Endometrial Neoplasms; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; Humans; Neoplasm Invasiveness; Progesterone; Reverse Transcriptase Polymerase Chain Reaction; Sucrose; Thromboplastin | 2005 |
The secretory leukocyte protease inhibitor gene is a target of epidermal growth factor receptor action in endometrial epithelial cells.
The over-expression of epidermal growth factor receptor (EGFR) and its ligands, epidermal growth factor (EGF) and transforming growth factor-alpha, is a common feature of epithelial carcinomas and correlates with neoplastic progression. Secretory leukocyte protease inhibitor (SLPI), a member of the Kazal superfamily of serine anti-proteases, induces proliferation and promotes malignancy of epithelial cells and is expressed at high levels in multiple tumor types. In the present study, we have demonstrated that EGF increases SLPI expression in the human endometrial epithelial cell line Ishikawa in a dose- and time-dependent manner. We have shown that this effect of EGF occurs, in part, at the level of the SLPI promoter and involves the MAP kinase signaling pathway. We have further shown that EGF promotion of cell proliferation, but not induction of cyclin D1 gene expression, involves SLPI. Our results suggest that the regulation of SLPI expression by EGFR ligand(s) may represent a 'feed-forward' mechanism by which the enhanced proliferative and migratory properties of EGFR over-expressing cancer cells are sustained. Increased SLPI expression is likely an important component of altered EGFR signaling in human tumors and may have significant therapeutic implications in cancer progression. Topics: Carcinoma; Cell Line, Tumor; Cell Proliferation; Dose-Response Relationship, Drug; Endometrial Neoplasms; Epidermal Growth Factor; Epithelial Cells; ErbB Receptors; Female; Gene Expression Regulation; Humans; Proteinase Inhibitory Proteins, Secretory; Proteins; Secretory Leukocyte Peptidase Inhibitor; Time Factors; Transfection; Transforming Growth Factor alpha | 2005 |
Tissue factor is regulated by epidermal growth factor in normal and malignant human endometrial epithelial cells.
Tissue Factor (TF), the initiator of the extrinsic coagulation cascade, is overexpressed in a variety of cancers. TF is also expressed in normal human endometrium but little is known about its expression or regulation in endometrial cancer. We demonstrate herein that TF is expressed in the endometrial adenocarcinoma cell line Ishikawa. Furthermore, epidermal growth factor (EGF) induces a rapid and sustained increase in TF expression. Estradiol and progesterone had no effect on basal or EGF-induced TF expression in Ishikawa cells. In contrast to the pronounced and sustained upregulation at the protein level, EGF treatment elicited only a modest and transient increase in TF mRNA levels. This activity corresponded to the response observed from an exogenous TF promoter construct. However, the induction of TF was abrogated by cycloheximide as well as actinomycin-D, inhibitors or protein- and mRNA-synthesis, respectively, demonstrating that EGF mediates its effect through activation of the TF gene. Fractionation experiments showed that EGF increases TF presence in caveolin-I containing membrane fractions. Coagulation and invasion assays were used to explore the physiological implications of TF regulation. The results demonstrate that EGF-mediated induction of TF increases the procoagulant activity and invasive potential of Ishikawa cells. Furthermore, immunocytochemistry confirmed that TF is regulated by EGF in primary cultures of normal endometrial epithelial cells and malignant tumor cells. In conclusion, EGF-mediated upregulation of TF results in accumulation of this glycoprotein in caveolae-like membrane fractions and increased coagulative and invasive potential. Our results suggest that TF may play an integral role in endometrial carcinogenesis. Topics: Adenocarcinoma; Anticoagulants; Blotting, Western; Cell Line, Tumor; Cells, Cultured; Coagulants; Cycloheximide; Dactinomycin; Detergents; Endometrial Neoplasms; Endometrium; Epidermal Growth Factor; Estradiol; Female; Gene Expression Regulation, Neoplastic; Glycoproteins; Humans; Immunohistochemistry; Nucleic Acid Synthesis Inhibitors; Octoxynol; Progesterone; Promoter Regions, Genetic; RNA, Messenger; Sucrose; Thromboplastin; Time Factors; Transfection; Ultracentrifugation; Up-Regulation | 2005 |
Regulation of signaling phosphoproteins by epidermal growth factor and Iressa (ZD1839) in human endometrial cancer cells that model type I and II tumors.
To understand how type I and II endometrial tumors uniquely respond to tyrosine kinase inhibitor treatments, we evaluated the signaling pathways of epidermal growth factor (EGF) receptor (EGFR) under the effects of EGF and Iressa (ZD1839, gefitinib) using Ishikawa H and Hec50co cells that model type I and II endometrial carcinomas, respectively. The cells were assayed for the expression of EGFR and both cell lines express an average of 100,000 EGFR per cell; however, Ishikawa H cells express higher levels of HER-2/neu compared with Hec50co cells (1.38 x 10(5) compared with 2.04 x 10(4), respectively). Using the Kinetworks multi-immunoblotting approach, which profiles 31 signaling phosphoproteins, the most striking result was that Hec50co cells show a higher number of basal phosphorylated sites compared with Ishikawa H cells. Furthermore, we identified targets of Iressa treatment in both cell lines. Iressa, at a dose of 1 micromol/L, blocked the autophosphorylation of EGFR in Ishikawa H and Hec50co cells with some distinctive effects on downstream effectors. Nevertheless, in both cell lines, EGF stimulated and Iressa blocked the major EGFR target mitogen-activated protein kinases extracellular signal-regulated kinase 1 and 2 equally. The high basal phosphorylation of numerous signaling molecules in Hec50co cells that were not inhibited by Iressa indicates that other growth factor pathways are active in addition to EGFR. We conclude that endometrial cancer cells that model type I and II carcinomas have the capacity to respond to EGFR inhibition as a therapeutic strategy; however, the response of the more aggressive type II tumors may be limited by the constitutive activation of other signaling pathways. Topics: Antineoplastic Agents; Blotting, Western; Cell Line, Tumor; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Flow Cytometry; Gefitinib; Humans; Phosphoproteins; Quinazolines; Signal Transduction | 2005 |
Quercetin regulates growth of Ishikawa cells through the suppression of EGF and cyclin D1.
Quercetin and other polyphenols have anti-carcinogenic and anti-tumorigenic activity in various organs, however, studies of this activity are lacking in endometrial cancer. We hypothesize that quercetin has anti-proliferative activity and the mechanisms of quercetin action may be through modulation of cell cycle and cell growth regulatory genes. To test this hypothesis, we treated endometrial cancer cells (Ishikawa cell line) with quercetin, and cell proliferation, expression of growth signal genes (EGF, VEGF, and TGF-alpha), cell cycle genes (p53, p21, p73, and cyclin D1), and apoptosis-related genes (bcl-2 and bax) were analyzed. Results of these experiments demonstrate that after a 7-day exposure to 1, 10 and 100 micro M of quercetin, growth of Ishikawa cells was inhibited by 3, 51 and 87%, respectively. The gene and protein expression data suggest that quercetin treatment (100 micro M) significantly decreased EGF and cyclin D1, whereas VEGF was up-regulated in Ishiwaka cell lines. Other genes such as TGF-alpha, p53, p21, p73, bcl-2 and bax were not significantly changed with quercetin treatment in Ishiwaka cell lines. The present study suggests that quercetin can suppress proliferation of Ishikawa cells through down-regulation of EGF and cyclin D1. Topics: Cyclin D1; Cyclin-Dependent Kinase Inhibitor p21; Cyclins; Endometrial Neoplasms; Endothelial Growth Factors; Epidermal Growth Factor; Female; Gene Expression Regulation; Humans; Intercellular Signaling Peptides and Proteins; Lymphokines; Quercetin; Tumor Cells, Cultured; Tumor Suppressor Protein p53; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors | 2003 |
Resveratrol suppresses growth of Ishikawa cells through down-regulation of EGF.
Resveratrol and other polyphenols have anti-carcinogenic and anti-tumorigenic activities in various carcinomas. However, such studies are limited in endometrial cancer. We hypothesize that resveratrol suppresses cancer growth through modulation of cell cycle and cell growth regulatory genes. To test this hypothesis, we treated endometrial cancer cells (Ishikawa cell line) with resveratrol (1, 10, 50 and 100 micro M) for 1, 3, 5 and 7 days, and analyzed for growth signal genes (EGF and VEGF), cell cycle regulatory genes (p53 and p21), and apoptosis-related genes (bcl-2 and bax). Results of these experiments demonstrate that after resveratrol treatment, the growth of Ishikawa cells was inhibited in a dose dependent manner. The gene and protein expression data suggest that resveratrol treatment significantly decreased EGF, whereas VEGF was up-regulated in Ishiwaka cell lines. Interestingly, protein expressions of p21 and Bax were decreased, even though their mRNA expressions did not show significant changes. The present study suggests that resveratrol can suppress proliferation of Ishikawa cells through down-regulation of EGF. Topics: Antineoplastic Agents, Phytogenic; Apoptosis; Blotting, Western; Cell Cycle; Cell Division; Cell Line, Tumor; Dose-Response Relationship, Drug; Down-Regulation; Endometrial Neoplasms; Epidermal Growth Factor; Female; Humans; Resveratrol; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Stilbenes; Time Factors | 2003 |
Immunohistochemical investigation of p-53, C-NEU and EGFR expression in HPV-related epidermoid endometrial carcinoma.
Epidermoid carcinoma (PSCC) of the endometrium is a rare form of endometrial cancer that constitutes about 0.1% of all malignant epithelial tumors of the uterus. The diagnosis of PSCC is based on strict criteria and is made in the absence of a glandular component of the tumor. Squamous cell carcinoma of the endometrium should enter the differential diagnosis in postmenopausal patients in the presence of atypical squamous cells in the uterine curettage, while the cervical biopsies are negative for malignancy. The presence of HPV should be investigated as well, so that its pathogenetic relation is clarified. While no significant relation was found to p-53, C-NEU and EGFR expression this investigation must be continued because. HPV may interact with tumor suppressor genes. Topics: Aged; Biomarkers, Tumor; Biopsy, Needle; Carcinoma, Squamous Cell; Endometrial Neoplasms; Epidermal Growth Factor; Fatal Outcome; Female; Genes, p53; Humans; Immunohistochemistry; Papillomavirus Infections; Proto-Oncogene Proteins; Sensitivity and Specificity; Tumor Virus Infections | 2002 |
Antiproliferative signaling of luteinizing hormone-releasing hormone in human endometrial and ovarian cancer cells through G protein alpha(I)-mediated activation of phosphotyrosine phosphatase.
The signaling pathway through which LHRH acts in endometrial and ovarian cancers is distinct from that in the anterior pituitary. The LHRH receptor interacts with the mitogenic signal transduction of growth factor receptors, resulting in down-regulation of expression of c-fos and proliferation. Only limited data are available on the cross-talk between LHRH receptor signaling and inhibition of mitogenic signal transduction. The present experiments were performed to analyze in endometrial and ovarian cancer cells: 1) whether mutations or splice variants of the LHRH receptor are responsible for differences in LHRH signaling, 2) the coupling of G protein subtypes to LHRH receptor, 3) the phosphotyrosine phosphatase (PTP) activation counteracting growth factor receptor tyrosine kinase activity. For these studies, the well characterized human Ishikawa and Hec-1A endometrial cancer cell lines and human EFO-21 and EFO-27 ovarian cancer cell lines were used, which express LHRH and its receptor. 1) Sequencing of the complementary DNA of the LHRH receptor from position 31 to position 1204, covering the complete coding region (position 56 to position 1042) showed that there are neither mutations nor splice variants of the LHRH receptor transcript in Ishikawa and Hec-1A endometrial cancer cells or in EFO-21 and EFO-27 ovarian cancer cells. 2) All analyzed cell lines except for the ovarian cancer cell line EFO-27 expressed both G proteins, alpha(i) and alpha(q), as shown by RT-PCR and Western blotting. In the EFO-27 cell line only G protein alpha(i), not G protein alpha(q), expression was found. Cross-linking experiments using disuccinimidyl suberate revealed that in the cell lines expressing G protein alpha(i) and G protein alpha(q), both G proteins coupled to the LHRH receptor. Inhibition of epidermal growth factor (EGF)-induced c-fos expression by LHRH, however, was mediated through pertussis toxin (PTX)-sensitive G protein alpha(i). Moreover, LHRH substantially antagonized the PTX-catalyzed ADP-ribosylation of G protein alpha(i). 3) Using a phosphotyrosine phosphatase assay based on molybdate-malachite green, treatment of quiescent EFO-21 and EFO-27 ovarian cancer cells and quiescent Ishikawa and Hec-1A endometrial cancer cells with 100 nM of the LHRH agonist triptorelin resulted in a 4-fold increase in PTP activity (P < 0.001). This effect was completely blocked by simultaneous treatment with PTX, supporting the concept of mediation through G protein alpha(i). As Topics: Adenosine Diphosphate Ribose; Alternative Splicing; Base Sequence; Cell Division; Endometrial Neoplasms; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression; Genes, fos; Gonadotropin-Releasing Hormone; GTP-Binding Protein alpha Subunits, Gi-Go; GTP-Binding Proteins; Humans; Molecular Sequence Data; Mutation; Ovarian Neoplasms; Pertussis Toxin; Phosphorylation; Phosphotyrosine; Protein Tyrosine Phosphatases; Receptors, LHRH; RNA, Messenger; Sequence Analysis, DNA; Signal Transduction; Tumor Cells, Cultured; Virulence Factors, Bordetella | 2001 |
Retinoic acid affects the EGF-R signaling pathway during differentiation induction of human endometrial adenocarcinoma cells.
We have shown that moderately differentiated endometrial adenocarcinoma (RL95-2) cells differentiate in response to retinoic acid treatment, illustrated by their reorganization of actin filaments and cell enlargement (Carter et al., Anticancer Res. 16, 17-24, 1996). Tyrphostin, an inhibitor of epidermal growth factor receptor (EGF-R)-associated protein tyrosine kinases, caused a dramatic reorganization of actin filaments in RL95-2 cells, similar to retinoic-acid-treated cells (Carter and Bellido, J. Cell. Physiol. 178, 320-332, 1999). We evaluated the possibility that the differentiating effects of retinoids are due to retinoic-acid-induced decreases in phosphorylation of EGF-R and changes in downstream effector proteins. Retinoic acid caused a decrease in tyrosine phosphorylation of EGF-R. Retinoic acid treatment induced a dramatic actin filament reorganization and cell enlargement. Treatment with EGF reversed this effect, because cells treated with retinoic acid followed by EGF only possessed disrupted actin aggregates and appeared small, thus resembling medium controls. Retinoic acid induced a relocalization and decrease in the amount of Shc protein, another actin-binding protein which is an adaptor protein for EGF-R signaling. In addition, retinoic acid induced a relocalization of gelsolin from the plasma membrane to the cytoplasm. Retinoic acid decreased cell detachment in detachment assays; one-half as many retinoic-acid-treated cells detached as in controls. These results are consistent with the idea that retinoic acid induces differentiation of RL95-2 cells by interfering with the EGF-R signaling pathway. Topics: Actin Cytoskeleton; Actins; Adaptor Proteins, Signal Transducing; Adaptor Proteins, Vesicular Transport; Adenocarcinoma; Cell Adhesion; Cell Differentiation; Cell Membrane; Cyclic AMP-Dependent Protein Kinases; Cytoplasm; Cytoskeleton; Endometrial Neoplasms; Enzyme Activation; Epidermal Growth Factor; ErbB Receptors; Female; Gelsolin; GRB2 Adaptor Protein; Humans; Isotretinoin; Membrane Proteins; Neoplasm Proteins; Phosphorylation; Protein Processing, Post-Translational; Proteins; Shc Signaling Adaptor Proteins; Signal Transduction; Src Homology 2 Domain-Containing, Transforming Protein 1; Tetradecanoylphorbol Acetate; Tretinoin; Tumor Cells, Cultured | 2000 |
Inhibition of human endometrial cancer cell growth in vitro and in vivo by somatostatin analog RC-160.
Our purpose was to investigate the effect of the somatostatin analog RC-160 on the growth of the HEC-1 human endometrial cancer cell line in vivo and in vitro.. Nude mice bearing subcutaneous implanted HEC-1 tumors were treated for 25 days with RC-160 (100 microgram/d) delivered by osmotic minipumps. In cultured HEC-1 cells radioreceptor assay of somatostatin was performed, and the expression of messenger ribonucleic acid for somatostatin receptor subtypes (somatostatin receptors 1-5) was analyzed by reverse transcription-polymerase chain reaction. The effects of RC-160 on epidermal growth factor-stimulated cell proliferation and tyrosine phosphorylation of epidermal growth factor receptor were examined by colorimetric assay and Western blotting, respectively.. The treatment with RC-160 resulted in a significant decrease in tumor volume, tumor weight, and serum insulin-like growth factor I levels compared with those values in control animals. The presence of high-affinity somatostatin binding sites and the expression of somatostatin receptor 2 and somatostatin receptor 3 messenger ribonucleic acid were demonstrated in HEC-1 cells by radioreceptor assay and reverse transcription-polymerase chain reaction, respectively. Epidermal growth factor-stimulated proliferation of HEC-1 cells was inhibited by RC-160 in a dose-dependent manner. Western blotting revealed that epidermal growth factor-induced tyrosine phosphorylation of epidermal growth factor receptor was inhibited by RC-160, which suggests that the direct inhibitory effect of RC-160 on HEC-1 cell growth might be mediated in part by interference with epidermal growth factor receptor phosphorylation.. These results indicate that somatostatin analog RC-160 inhibits the growth of HEC-1 human endometrial cancer cells, thus implying its potential clinical utility in treating endometrial cancer. Topics: Animals; Antineoplastic Agents; Cell Division; Endometrial Neoplasms; Epidermal Growth Factor; Female; Humans; Mice; Mice, Inbred BALB C; Mice, Nude; Neoplasm Transplantation; Radioligand Assay; Receptors, Somatostatin; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Somatostatin | 1999 |
Biological implications of growth factors on the mechanism of invasion in gynecological tumor cells.
We investigated the effects of epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-alpha) on migration, invasion and proteinase expression of gynecological cultured cancer cells (SKG-IIIb cervical squamous cell carcinoma, OMC-4 cervical adenocarcinoma, SNG-M endometrial adenocarcinoma and OMC-3 ovarian adenocarcinoma), and whether these growth factors affect thymidine phosphorylase/platelet-derived endothelial cell growth factor expression of tumor cells. Tumor cell migration along a gradient of substratum-bound fibronectin and invasion into reconstituted basement membrane were stimulated by 0.1-10 nM EGF and TGF-alpha in a concentration-dependent manner. The zymography of tumor-conditioned medium showed that the treatment of tumor cells with EGF and TGF-alpha resulted in the increase of type IV collagenases, stromelysin and urokinase-type plasminogen activator which was partly confirmed by immunoblot analysis. The expression of thymidine phosphorylase/platelet-derived endothelial cell growth factor which has angiogenic activity, was also upregulated by these growth factors. These results suggest that EGF and TGF-alpha act as positive regulators on the invasion process of gynecological tumor cells which may be associated with their stimulatory action on the motility of tumor cells, the expression of proteinases secreted by tumor cells and the angiogenic phenotype. Topics: Adenocarcinoma; Carcinoma, Squamous Cell; Cell Movement; Endometrial Neoplasms; Epidermal Growth Factor; Female; Genital Neoplasms, Female; Humans; Metalloendopeptidases; Neoplasm Invasiveness; Ovarian Neoplasms; Thymidine Phosphorylase; Transforming Growth Factor alpha; Tumor Cells, Cultured; Urokinase-Type Plasminogen Activator; Uterine Cervical Neoplasms | 1999 |
Abnormal expression of cripto and p53 protein in endometrial carcinoma and its precursor lesions.
Many oncogenes and tumor suppressor genes are involved in multistep carcinogenesis. Cripto is an epidermal growth factor (EGF) related gene which shares homology with EGF and TGFalpha. The aim of this study was to evaluate the role of abnormal p53 and cripto oncogene expression in endometrial carcinogenesis and progression using a hyperplasia carcinoma sequence model. Ninety-six primary endometrial adenocarcinomas and 30 hyperplastic tissues of which 7 were atypical (AH), were immunohistochemically examined for the presence of cripto and abnormal p53 protein. Immunopositivity was compared in hyperplastic and carcinoma tissues and analysed for conventional clinicopathological prognostic variables such as grade, depth of myometrial invasion, lymphovascular invasion, lymph node metastases and clinical stage. Cripto immunoreactivity was strong in most cases of AH, and endometrial carcinomas revealed 71% overall and 41% strong positivity, while hyperplasias without atypia were weakly stained. There was no correlation between cripto expression and clinicopathological prognosticators. Abnormal p53 was not observed in hyperplasias but AH and carcinomas expressed 14% and 25% overall positivity, respectively. There was a statistically significant correlation between the stage of the disease and abnormal p53 accumulation. Our results suggest that both cripto and p53 may play a role in endometrial carcinogenesis while abnormal p53 expression is an important parameter for disease progression. Topics: Carcinoma; Disease Progression; Endometrial Hyperplasia; Endometrial Neoplasms; Epidermal Growth Factor; Female; GPI-Linked Proteins; Humans; Immunohistochemistry; Intercellular Signaling Peptides and Proteins; Membrane Glycoproteins; Neoplasm Proteins; Tumor Suppressor Protein p53 | 1998 |
Oncogenic Ras modulates epidermal growth factor responsiveness in endometrial carcinomas.
Since the majority of endometrial carcinomas do not contain any detectable ras mutations, the precise contribution of aberrant Ras function, if any, to endometrial carcinoma development remains to be determined. Since there is considerable evidence that Ras transformation is associated with a decreased requirement for growth factors, we compared the growth response of endometrial carcinoma cells harbouring wild-type (Ishikawa cells) or mutated (HHUA cells) K-ras to epidermal growth factor (EGF). K-ras mutation did not significantly affect the level of the EGF receptor (EGFR) expressed in these carcinoma cells. EGF could stimulate the growth of Ishikawa, but not HHUA cells. Furthermore, EGF caused elevation of Ras-GTP levels in Ishikawa, but not HHUA cells. However, the introduction of mutated, but not normal, K-ras into Ishikawa cells rendered them non-responsive to EGF growth stimulation. Thus, the presence of mutated K-ras alone modulated the growth response of endometrial carcinoma cells to EGF. An inhibitor of the EGFR tyrosine kinase activity could prevent soft agar colony formation of Ishikawa cells, but not HHUA or mutant K-ras(12V)-transfected Ishikawa cells. Taken together, these results suggest that mutated K-ras causes a loss of responsiveness to EGF stimulation and that EGFR function is dispensable for the growth of mutant Ras-positive endometrial carcinoma cells. Topics: Apoptosis; Cell Division; Endometrial Neoplasms; Enzyme Inhibitors; Epidermal Growth Factor; ErbB Receptors; Female; Genes, ras; Humans; Hydroquinones; Mutation; Signal Transduction; Transforming Growth Factor alpha; Tumor Cells, Cultured | 1998 |
Transforming growth factor-alpha and insulin-like growth factor-I, but not epidermal growth factor, elicit autocrine stimulation of mitogenesis in endometrial cancer cell lines.
Endometrial carcinoma cell lines were evaluated for epidermal growth factor (EGF), transforming growth factor-alpha (TGF-alpha), and insulin-like growth factor I (IGF-1) production and for autocrine stimulation.. Conditioned, serum-free media (CM) from cell lines RL95-2, KLE, HEC, and Ishikawa (ISH) were concentrated radioimmunoassayed (RIA). Samples for the IGF-1 assay were extracted with acid-ethanol to remove IGF-1 binding protein. Polymerase chain reaction (PCR) was used to validate the presence of mRNA for growth factors and receptors. Cells were incubated with Ab528, an antibody blocking EGF receptors, and alphaIR3, an antibody blocking IGF-1 receptors. Proliferation was quantified using [3H]thymidine incorporation.. TGF-alpha was detected in CM: RL95-2 (0.4 +/- 0.001 ng/ml), KLE (0.7 +/- 0.003 ng/ml), HEC (0.8 +/- 0.01 ng/ml), ISH (1.2 +/- 0.05 ng/ml). No EGF was detected in CM. In extracted samples, IGF-1 was detected in CM: RL95-2 (0.8 +/- 0. 03 ng/ml), KLE (1.25 +/- 0.02 ng/ml), HEC (1.6 +/- 0.01 ng/ml), ISH (1.6 +/- 0.08 ng/ml). Unconditioned media served as the control. EGF, TGF-alpha, and IGF-1 mRNA was identified in all cell lines, as was the mRNA for EGF and IGF-1 receptors. Incubation with Ab528 or alphaIR3 resulted in significant inhibition of DNA synthesis in HEC 1A, KLE, and ISH. No inhibition was detected in the RL95-2 cell line. A control antibody did not inhibit the cell lines.. Autocrine production and stimulation of endometrial carcinoma cell lines by TGF-alpha and IGF-1 are demonstrated in three of four endometrial cancer cell lines. No measurable EGF was produced by any of the cell lines. Topics: Autocrine Communication; Cell Division; Culture Media, Conditioned; Culture Media, Serum-Free; DNA, Neoplasm; Endometrial Neoplasms; Epidermal Growth Factor; Female; Humans; Insulin-Like Growth Factor I; Neoplasm Proteins; Polymerase Chain Reaction; RNA, Messenger; Transforming Growth Factor alpha; Tumor Cells, Cultured | 1998 |
Role of estrogen and estrogen-related growth factor in the mechanism of hormone dependency of endometrial carcinoma cells.
The role of estrogen and estrogen-related growth factors in the mechanism of hormone dependency of endometrial adenocarcinoma cells was investigated. The proliferation of hormone-responsive human endometrial adenocarcinoma cells (Ishikawa cells), which possess both estrogen and progesterone receptors, was optimally stimulated by 10 nM estradiol. Both transforming growth factor (TGF)-alpha and epidermal growth factor (EGF), added to the culture media, stimulated the proliferation of Ishikawa cells in a dose-dependent manner. Anti-TGF-alpha antibody completely eliminated the stimulatory effects of TGF-alpha. Anti-EGF receptor antibody inhibited the proliferation of these cells. The production of TGF-alpha into culture media was 5-40 pg/10 cells/24 h in 9 human endometrial adenocarcinoma cells. Ten nanomoles of estradiol increased the TGF-alpha production of Ishikawa cells by approximately 2.5-fold of the control level. In contrast, the production of TGF-alpha in hormone-unresponsive HEC-50 cels was not influenced by estradiol. C-erbB-2 oncoprotein expression of human endometrial adenocarcinoma cells, detected by both immunocytochemical staining and Western blot analysis, was associated with the tumor grade of the original tumor tissues. Ten nanomoles of estradiol clearly increased the c-erbB-2 oncoprotein levels at an optimal incubation period of 72 h, whereas estradiol did not affect the expression in HEC-50 cells. Topics: Adenocarcinoma; Blotting, Western; Dose-Response Relationship, Drug; Endometrial Neoplasms; Epidermal Growth Factor; Estrogens; Female; Humans; Immunohistochemistry; Neoplasms, Hormone-Dependent; Receptor, ErbB-2; Time Factors; Transforming Growth Factor alpha; Tumor Cells, Cultured | 1998 |
Transforming growth factor-beta negatively modulates proliferation and c-fos expression of the human endometrial adenocarcinoma cell line HEC-1-A.
Growth factor regulation of normal and cancerous cell proliferation has been well-documented and may be mediated by proto-oncogene activity. The purpose of this study was to assess changes in proliferation and mitogen-induced c-fos mRNA expression of an endometrial carcinoma cell line, HEC-1-A, in response to TGF-beta, a potent growth-inhibitory peptide. HEC-1-A cells were incubated in the presence or absence of TGF-beta. Mitogen-stimulated cells were additionally treated with epidermal growth factor (EGF). Changes in proliferation were measured by [3H]thymidine uptake assays. Alterations in EGF-induced c-fos expression following TGF-beta pretreatment were assessed by Northern blot using a 32P-labeled human c-fos probe. Finally, chloramphenicol acetyltransferase assays were performed to evaluate c-fos promoter activity in response to treatment conditions. Basal and EGF-stimulated proliferation was inhibited by TGF-beta in a dose- and time-dependent manner. TGF-beta also reversibly decreased EGF-induced c-fos mRNA expression in a dose- and time-dependent manner. Sequences in the c-fos promoter that were stimulated by EGF showed suppressed activity when preincubated with TGF-beta. These results show that TGF-beta negatively modulates EGF-induced c-fos expression, which may be related to the observed inhibition of carcinoma cell proliferation. Topics: Adenocarcinoma; Blotting, Northern; Cell Division; DNA, Neoplasm; Dose-Response Relationship, Drug; Drug Interactions; Endometrial Neoplasms; Epidermal Growth Factor; Female; Gene Expression Regulation, Neoplastic; Genes, fos; Humans; Promoter Regions, Genetic; Proto-Oncogene Mas; Proto-Oncogene Proteins c-fos; RNA, Messenger; Time Factors; Transfection; Transforming Growth Factor beta; Tritium; Tumor Cells, Cultured | 1997 |
Epidermal growth factor and sex steroids dynamically regulate a marker of endometrial receptivity in Ishikawa cells.
The factors regulating human endometrial receptivity remain poorly understood. The alpha v beta 3 integrin cell adhesion molecule appears to be regulated in the human endometrium, appearing on postovulatory days 5-6, corresponding to the time of initial embryo attachment. This integrin has been extensively studied as a potential marker of endometrial receptivity and is aberrantly expressed in the endometrial epithelium of some infertile women. Ishikawa cells are a well differentiated endometrial adenocarcinoma cell line that maintain functional estrogen and progesterone receptors and are a useful model to study steroid-mediated events in human endometrial epithelium. This cell line expresses most of the normal endometrial epithelial integrins, including the alpha v beta 3 vitronectin receptor. The regulation of this integrin was studied with fluorescence immunocytochemistry, flow cytometry, and Northern blot analysis. Estrogen with or without progesterone treatment down-regulates alpha v beta 3 in this cell line. Several growth factors, including epidermal growth factor and the closely related transforming growth factor-alpha significantly increase the expression of this integrin. We conclude that endometrial differentiation is influenced by both steroid hormones and growth factors. The alpha v beta 3 integrin appears to be an excellent marker to study the molecular events leading to the establishment of uterine receptivity and successful implantation. Topics: Adenocarcinoma; Biomarkers; Blotting, Northern; Cytokines; Dose-Response Relationship, Drug; Endometrial Neoplasms; Epidermal Growth Factor; Estrogens; Female; Flow Cytometry; Fluorescent Antibody Technique, Indirect; Humans; Progesterone; Receptors, Vitronectin; Tumor Cells, Cultured | 1997 |
Epidermal growth factor activates protein kinase C in the human endometrial cancer cell line HEC-1-A.
Previous studies from this laboratory have shown that epidermal growth factor (EGF) and the tumor promoter, phorbol myristate acetate (PMA), are mitogenic in the endometrial cancer cell line HEC-1-A. Since the effects of EGF have been shown to be mediated by the protein kinase C (PKC) pathway transduction system, we examined the possibility that the EGF-responsive signal in the endometrial cancer cell line HEC-1-A involves protein kinase C activation. HEC-1-A cells were grown to confluency in 100-mm dishes and maintained in a serum-free medium for 24 hr prior to treatment. The cells were treated with EGF at varying time intervals (0.25 to 60 min) and concentrations (0.1 to 200 ng/ml). The cells were then lysed, homogenized, and centrifuged at 105,000g for 1 hr at 4 degrees C. The supernatant was chromatographed on DEAE-Sephacel columns. The membranous pellet was resuspended in 5 ml of lysis buffer containing 1% Nonidet P-40 and also chromatographed on DEAE-Sephacel columns separately. The eluates were collected and assayed for protein kinase C activity by determining the amount of 32P transferred from [gamma-32P]ATP onto histones in the presence of the phospholipids, phosphatidylserine, and diolein. Our results show that the cytoplasmic and membrane fraction of the HEC-1-A cell line contained phosphotransferase activity which displayed kinetic characteristics typical of the protein kinase C enzyme. The optimal incubation time for protein kinase C activation in the cytosol by EGF was 5 min (30-fold stimulation). The protein kinase C activity was increased when the cell lines were incubated with increasing concentrations of EGF. Enzyme saturation was seen at a concentration of 10 ng/ml of EGF (4.5-fold stimulation). Western blot analysis confirmed the presence of the PKC enzyme in the cytosol and membranes of our cancer cell line. These results suggest that EGF, at least partially, exerts its effects on the endometrial adenocarcinoma cell line by activating protein kinase C through increased breakdown of phosphatidyl inositol (PI). The PI cascade appears to be an important signal transduction system mediating the growth stimulatory effects of EGF on endometrial carcinoma. Topics: Blotting, Western; Cell Membrane; Cytosol; Diglycerides; Down-Regulation; Endometrial Neoplasms; Enzyme Activation; Epidermal Growth Factor; Female; Humans; Hypoglycemic Agents; Insulin; Insulin-Like Growth Factor I; Insulin-Like Growth Factor II; Kinetics; Protein Kinase C; Stimulation, Chemical; Tumor Cells, Cultured | 1997 |
Expression of epidermal growth factor family proteins and epidermal growth factor receptor in human endometrium.
The biological significance of epidermal growth factor (EGF)-related proteins in the development and progression of endometrioid endometrial carcinoma was studied. Expression of EGF-related proteins including EGF, transforming growth factor-alpha (TGF-alpha), cripto (CR), amphiregulin (AR), and EGF receptor (EGFR) were immunohistochemically examined. Results were then correlated with clinicopathologic findings and steroid receptor status in 12 specimens with normal endometrium, 13 with endometrial hyperplasia, and 40 with endometrioid endometrial carcinoma. EGFR, EGF, TGF-alpha, and CR immunoreactivities were observed in 58.3%, 66.7%, 91.6%, and 66.7% of normal endometrial specimens; 100%, 15.4%, 100%, and 30.8% of endometrial hyperplasia specimens; and 67.5%, 32.5%, 65.0%, and 65.0% of endometrial carcinoma specimens, respectively. AR immunoreactivity was not observed in any of the normal, hyperplastic, or neoplastic endometrium. The presence or absence of EGFR or TGF-alpha in endometrial carcinoma correlated with surgical stage, depth of myometrial invasion, and findings from peritoneal washing cytology. EGF expression significantly correlated with the age of the patients and that of CR with surgical stage and peritoneal washing cytological findings. There was a significant correlation between EGFR and TGF-alpha expression, and between EGF and TGF-alpha. Co-expression of EGFR and TGF-alpha, EGFR and CR, and TGF-alpha and CR in carcinoma specimens significantly correlated with advanced surgical stage, deeper myometrial invasion, and positive peritoneal washing cytology. In normal as well as hyperplastic endometrium, endometrial glands immunohistochemically positive for TGF-alpha were generally positive for ER, but in poorly differentiated endometrial carcinoma, cells positive for TGF-alpha tended to be negative for ER. The results of the present study show that among EGF-related proteins, expression of TGF-alpha and CR seem to be associated with the progression of human endometrioid endometrial carcinoma. Additionally, expression of TGF-alpha became increasingly estrogen independent with increasing histological carcinoma grades. Topics: Adult; Amphiregulin; Carcinoma, Endometrioid; EGF Family of Proteins; Endometrial Hyperplasia; Endometrial Neoplasms; Endometrium; Epidermal Growth Factor; ErbB Receptors; Female; Glycoproteins; GPI-Linked Proteins; Growth Substances; Humans; Intercellular Signaling Peptides and Proteins; Membrane Glycoproteins; Middle Aged; Neoplasm Proteins; Neoplasm Staging; Receptors, Estrogen; Receptors, Progesterone; Receptors, Steroid; Retrospective Studies; Transforming Growth Factor alpha | 1996 |
Effects of sex steroids and growth factors on migration and invasion of endometrial adenocarcinoma SNG-M cells in vitro.
Biological effects of sex steroids (estradiol-17beta, E2; progesterone, P; medroxyprogesterone acetate, MPA; Danazol, DZ) and growth factors (epidermal growth factor, EGF; transforming growth factor, TGF-alpha,beta) on migration and invasion of endometrial adenocarcinoma SNG-M cells were investigated by haptotactic migration and haptoinvasion assay. The enzymatic degradation of the extracellular matrix by tumor cells was also examined. Tumor cell migration along a gradient of substratum-bound fibronectin was inhibited by 0.1-10 microM MPA and DZ, but promoted by 0.1-10 nM EGF and TGF-alpha in a concentration-dependent manner. E2, P and TGF-beta did not have any effect on the motility of tumor cells. These effects were also confirmed by wound assay. The invasive activity of SNG-M cells into reconstituted basement membrane (Matrigel) was inhibited by the presence of 0.1-10 microM MPA and DZ, but promoted by 0.1-10 nM EGF and TGF-alpha in a concentration-dependent manner. E2, P and TGF-beta did not have any effect on tumor cell invasion. The zymography of tumor-conditioned medium showed that the treatment of SNG-M cells with EGF and TGF-alpha resulted in the increase of the 68, 72 and 92 kDa type IV collagenases (matrix metalloproteinase, MMP-2 and 9). Sex steroids and TGF-beta did not have significant effects on MMP-2 and 9. Stromelysin (MMP-3), also secreted by SNG-M cells, was not affected by sex steroids and growth factors. These results suggest that EGF and TGF-alpha act as positive regulators on the invasion process of endometrial adenocarcinoma cells, which may partly be associated with the induction of type IV collagenase secretion by tumor cells. The inhibitory effects of MPA and DZ on tumor cell invasion may depend at least partly on their inhibitory action on the motility of tumor cells. Topics: Adenocarcinoma; Caseins; Cell Division; Cell Movement; Culture Media, Conditioned; Danazol; Endometrial Neoplasms; Epidermal Growth Factor; Estrogen Antagonists; Female; Gelatin; Humans; Neoplasm Invasiveness; Progestins; Transforming Growth Factor alpha; Tumor Cells, Cultured | 1996 |
Promoter-specific activation of mouse lactoferrin gene by epidermal growth factor involves two adjacent regulatory elements.
The mouse lactoferrin gene responded to forskolin, 12-O-tetradecanoyl phorbol-13-acetate, and epidermal growth factor (EGF) stimulation via two adjacent enhancer elements, the cAMP response element (CRE) and EGF response element (EGFRE), collectively referred to as the mitogen response unit. In this report, we examined the minimal promoter and enhancer elements of the mouse lactoferrin gene that are required for EGF-induced transcriptional activation. We found that the CRE and noncanonical TATA box (ATAAA) are the minimal promoter elements for basal activity of the chloramphenicol acetyltransferase (CAT) reporter construct whereas the EGFRE is needed for an additional activity induced by EGF in transiently transfected human endometrial carcinoma RL95-2 cells (RL95-2). The EGFRE, however, did not function in heterologous promoters [SV 40 and thymidine kinase (TK)]. Therefore, EGF-stimulated lactoferrin gene activity is promoter specific in RL95-2 cells. In transiently transfected cells, EGF and forskolin showed synergistic effects on the CAT reporter that contained both response elements. Mutation made at either element or insertion of extra nucleotides between the two elements severely affected EGF-stimulated activity. Nuclear protein prepared from RL95-2 cells formed three complexes (A, B, and C) with the oligonucleotides containing both EGFRE and CRE in electrophoretic mobility shift assay. A new complex (E) was detected with the nuclear protein of EGF-treated cells. By oligonucleotide competition experiments, we demonstrated that the complex E was generated by protein bound to CRE. EGF-induced binding activity could be abolished by calf intestinal alkaline phosphatase but not by the protein synthesis inhibitor, cycloheximide. Therefore, binding of a preexisting phosphoprotein to the CRE region could be one of the requirements for EGF-induced mouse lactoferrin gene promoter activity. Topics: Animals; Binding Sites; Carcinoma; Electrophoresis; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Gene Expression Regulation; Humans; Lactoferrin; Mice; Nuclear Proteins; Promoter Regions, Genetic; Receptors, Cyclic AMP; Recombinant Proteins; TATA Box; Transfection; Tumor Cells, Cultured | 1996 |
Regulation of the promoter of the human corticotropin-releasing hormone gene in transfected human endometrial cells.
Corticotropin-releasing hormone (CRH) is expressed in several peripheral tissues, including normal epithelial cells of the human and rodent uterus. However, the biological role of endometrial CRH is known in neither species. As a first step to clarify this role, we studied the regulation of CRH promoter in endometrial cells. We performed homologous transfection experiments in Ishikawa cells, a human endometrial cell line, using a 0.9-kb fragment of the 5'-flanking region of the human CRH gene coupled to luciferase. Transfected cells were exposed for 18 h to 8-bromo cyclic adenosine monophosphate, forskolin, epidermal growth factor, steroids (estradiol, progesterone, and the synthetic glucocorticoid dexamethasone and their antagonists), and prostaglandin E2; then the activity of the luciferase reporter was determined in the cell lysates. We found that the activity of the 5'-flanking region of the CRH gene was stimulated by cyclic adenosine monophosphate and epidermal growth factor and inhibited in a receptor-mediated, dose-dependent fashion by estradiol and dexamethasone. The antiglucocorticoid RU 486 acted as a glucocorticoid agonist, suppressing the CRH gene activation, while progesterone was devoid of any activity. Prostaglandin E2 stimulated the CRH activation, and the prostanoid inhibitor indomethacin suppressed it, most probably by inhibiting endogenous prostaglandins. These findings suggest that endometrial CRH gene expression may be under the negative control of estrogens and glucocorticoids and under the positive control of prostaglandin E2. Topics: Adenocarcinoma; Corticotropin-Releasing Hormone; Cyclic AMP; Dexamethasone; Dinoprostone; Endometrial Neoplasms; Endometrium; Epidermal Growth Factor; Estradiol; Female; Gene Expression Regulation; Humans; Indomethacin; Luciferases; Mifepristone; Promoter Regions, Genetic; Recombinant Fusion Proteins; Transcriptional Activation; Transfection; Tumor Cells, Cultured | 1996 |
Immunohistochemical study of estradiol, epidermal growth factor, transforming growth factor alpha and epidermal growth factor receptor in endometrial neoplasia.
In a total of 113 cases of endometrial neoplasm, we studied the immunohistochemical expression of estradiol (E2), epidermal growth factor (EGF), transforming growth factor alpha (TGFalpha), and epidermal growth factor receptor (EGFR). Positive immunoreactivity of E2 was found in 61% of the neoplasms. E2 immunoreactivity correlated well with high histologic grade and early clinical stage. Positive immunoreactivity for EGF or EGFR was found in 25.6% or 53.1% of the neoplasms, respectively. However, this was unrelated to histologic grade or clinical stage. On the other hand, TGFalpha immunoreactivity was found in 67% of endometrial neoplasias and was correlated with poor histologic grade and advanced stage. Contingency tables indicated a significant negative association between the status of E2 and that of TGFalpha. Simultaneous expression of E2, EGF and EGFR, or E2, TGFalpha and EGFR was found in 6.8% and 15.9% of endometrial carcinomas, respectively. These results suggest that a predominant number of endometrial carcinomas escape autocrine/paracrine growth regulation by EGF and E2 or TGFalpha and E2, and that TGFalpha may be involved in the progression of endometrial carcinoma. Topics: Adult; Aged; Aged, 80 and over; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Estradiol; Female; Frozen Sections; Humans; Immunohistochemistry; Medroxyprogesterone Acetate; Middle Aged; Neoplasm Staging; Paraffin Embedding; Transforming Growth Factor alpha; Tumor Cells, Cultured | 1996 |
Estrogen induces c-Ha-ras expression via activation of tyrosine kinase in uterine endometrial fibroblasts and cancer cells.
Endometrial fibroblasts derived from uterine endometrium as controls and endometrial cancer cells (Ishikawa and HHUA cells) were used to analyze the manner of induction of c-Ha-ras transcripts in endometrial cancers, some of which are estrogen-dependent in growth. Estrogen increased c-Ha-ras expression and tyrosine kinase (TK) activity in fibroblast and Ishikawa cells, but not in HHUA cells. Progesterone diminished c-Ha-ras expression and tyrosine kinase (TK) activity induced by estradiol in the fibroblasts, but not in Ishikawa cells, which persistently overexpressed c-Ha-ras. In these cells, epidermal growth factor (EGF) increased c-Ha-ras expression as did estradiol. Pretreatment with tyrphostin, an inhibitor of TK, abolished estrogen-inducible overexpression of c-Ha-ras. The combination of both estradiol and EGF at maximum effective concentration exerted no additive or synergistic effect on induction of c-Ha-ras expression. In conclusion, persistent activation of TK might lead to overexpression of c-Ha-ras in some endometrial cancer cells under estrogen predominant milieu, which might be associated with the transformation or growth potential. Topics: Adenocarcinoma; Adult; Base Sequence; Carcinoma; Endometrial Neoplasms; Endometrium; Enzyme Activation; Enzyme Inhibitors; Epidermal Growth Factor; Estradiol; Female; Fibroblasts; Gene Expression Regulation; Gene Expression Regulation, Neoplastic; Humans; Molecular Sequence Data; Progesterone; Protein-Tyrosine Kinases; Proto-Oncogene Proteins p21(ras); RNA, Messenger; Tumor Cells, Cultured | 1995 |
[Expression of epidermal growth factor, transforming growth factor-alpha and epidermal growth factor receptor messenger RNA in human endometrium and endometrial carcinoma].
To study the regulation of c-erbB, EFG and TGF-alpha in endometrium, we examined the expression of their mRNA in 5 normal endometrial and 5 endometrial carcinoma tissues by Northern blot and reverse transcription-polymerase chain reaction (RT-PCR). By Northern blot with 10 micrograms of total RNA, c-erbB mRNA was not detected in the normal endometrium, but it was detected in 3 samples of endometrial carcinoma tissues. On the other hand RT-PCR identified c-erbB mRNA in all the normal endometrium and endometrial carcinoma tissues by amplifying cDNA derived from c-erbB mRNA. EGF mRNA was detected by RT-PCR in normal endometrium except in the early follicular phase. It was detected in all cases of endometrial carcinoma tissues. TGF-alpha mRNA was also detected in all the normal and endometrial carcinoma tissues by RT-PCR. Our study suggests that an autocrine/paracrine mechanism of EGF may regulate the endometrial cycle. Because some endometrial carcinoma tissues express the c-erbB mRNA much more than normal endometrium, disruption of the autocrine/paracrine mechanism may trigger subsequent endometrial carcinogenesis. Topics: Adenocarcinoma; Adult; Base Sequence; Blotting, Northern; Endometrial Neoplasms; Endometrium; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Middle Aged; Molecular Sequence Data; Polymerase Chain Reaction; RNA, Messenger; RNA, Neoplasm; Transforming Growth Factor alpha | 1995 |
Comparative analysis of the sensitivity of endometrial cancer cells to epidermal growth factor and steroid hormones.
Epidermal growth factor (EGF) and EGF-regulated processes play an important role in steroid signal transduction. Comparative analysis of EGF and steroid receptor expression and the sensitivity of early stages of proliferation induction, such as activation of phospholipid turnover to EGF and steroids, may provide a useful new approach to characterizing the sensitivity of endometrial cancer to hormone therapy.. Progesterone (PR), estradiol (ER), and EGF receptor (EGFR) content was measured by radioligand competitive methods in surgically excised tumors from 26 patients with primary endometrial cancer. In short term cell cultures isolated from 11 of these tumors, the influence of a 10-minute treatment with 10(-8)M EGF either alone or combined with 10(-8)M progesterone on 32P-incorporation into phospholipids was studied. Phospholipids were fractionated by thin-layer chromatography and were located by autoradiography, and quantification of the labeled compounds was made by densitometric scanning of the autoradiograms.. Epidermal growth factor receptor was found in 15 of 26 (58%) endometrial cancer samples. Eighty-two percent of the tumors studied contained PR, and 81% contained ER. No significant correlations were revealed between EGFR and ER/PR status or concentration. Epidermal growth factor stimulated 32P-incorporation by more than 120% of the control level in five of seven EGFR-positive and in one of four EGFR-negative endometrial cancer samples. An inverse relationship was revealed between EGFR content and the percentage of EGF-induced stimulation of phospholipid turnover in endometrial cancer cells (r = -0.6; P = 0.15) and between EGFR content in EGFR-positive samples and the extent of progesterone suppression of EGF-induced turnover (r = -0.77; P = 0.04).. Determination of EGF sensitivity on a receptor and a functional level may provide important additional information about the hormonal sensitivity of endometrial cancer. Topics: Adult; Aged; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Gonadal Steroid Hormones; Humans; Middle Aged; Receptors, Estradiol; Receptors, Progesterone | 1995 |
[Molecular endocrinology of growth factors].
Topics: Animals; Endometrial Neoplasms; Epidermal Growth Factor; Estrogens; Female; Humans; Somatomedins; Uterus | 1995 |
Expression of insulin-like growth factors and their binding proteins in the estrogen responsive Ishikawa human endometrial cancer cell line.
In uterine tissue, estrogen regulates various components of the insulin-like growth factor system; however, there are few suitable in vitro systems to examine these effects. Here we have examined the effects of 17-beta estradiol (E2) on expression and synthesis of insulin-like growth factors (IGF-I and IGF-II) and the insulin-like growth factor binding proteins (IGFBPs) by Ishikawa human endometrial cancer cells. Using a semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) assay, we demonstrated that both E2 and 4-hydroxy-tamoxifen (OHT) enhanced IGF-I expression but had no effect on IGF-II expression. The pure antiestrogen ICI 182,780 had no effect on IGF-I expression and partially blocked the E2 and OHT effect on IGF-I expression. The effect of epidermal growth factor (EGF), which is able to mimic some of the effects of E2 in Ishikawa cells and uterine tissue, was also examined. EGF, unlike E2, did not increase IGF-I expression but rather resulted in a significant decrease in IGF-I messenger RNA (mRNA) levels. EGF also resulted in a small, nonsignificant increase in IGF-II mRNA levels. IGFBP-3, -5, and -6 mRNAs were detected by Northern blot analyses of Ishikawa cells RNA. However, only IGFBP-3 was consistently detected by ligand blotting of conditioned medium. E2 had no significant effect on expression of any of the binding proteins, whereas EGF increased IGFBP-5 mRNA levels. These data provide the first in vitro demonstration of regulation of IGF-I expression by E2. The Ishikawa cell line may provide a useful model to further investigate the molecular mechanisms underlying E2 regulation of IGF-I expression. Furthermore, we have demonstrated a clear dissociation of the effects of E2 and EGF on IGF-I expression in this cell line. Topics: Base Sequence; Carrier Proteins; Endometrial Neoplasms; Epidermal Growth Factor; Estradiol; Estrogen Antagonists; Female; Gene Expression; Humans; Insulin-Like Growth Factor Binding Proteins; Insulin-Like Growth Factor I; Insulin-Like Growth Factor II; Molecular Probes; Molecular Sequence Data; Polymerase Chain Reaction; RNA, Messenger; Somatomedins; Tamoxifen; Transcription, Genetic; Tumor Cells, Cultured | 1994 |
The epidermal growth factor receptor in the human endometrial adenocarcinoma cell line HEC-1-B.
The binding characteristics and steroidal regulation of the EGF receptor were investigated in the human endometrial adenocarcinoma cell line HEC-1-B. The cell line was shown to possess a single, high affinity binding site for epidermal growth factor receptor (EGF) with a Kd of 3.09 +/- 1.39 nM (mean +/- SD, n = 6) and binding of 845 +/- 311 fmol/mg protein (mean +/- SD, n = 6). The protein kinase C activator, phorbol 12-myristate, 13-acetate (PMA) increased the Kd of the EGF receptor in a dose dependent manner (PMA: 0, 1, 10, 100 nM; Kd: 4.1, 5, 10, 50 nM, respectively). The effect of PMA (10 nM) was overcome by preincubating the cells with the protein kinase C inhibitor staurosporine (1 microM) prior to the addition of PMA. The effect of the ovarian steroids oestradiol and progresterone on EGF receptor accumulation was studied by pretreating the cells for 6 days with oestradiol or progesterone in phenol red free DMEM:F12, 1:1 supplemented with 5% charcoal stripped fetal calf serum. Both steroids were shown to increase EGF receptor number with a maximum 5- and 7-fold increase in the presence of 1 nM oestradiol or 1 microM progresterone, respectively. The study demonstrates the presence of a high affinity binding site for EGF in HEC-1-B cells which is regulated by oestradiol and progresterone. Topics: Adenocarcinoma; Alkaloids; Binding Sites; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Estradiol; Female; Humans; Kinetics; Progesterone; Protein Kinase C; Staurosporine; Tetradecanoylphorbol Acetate; Tumor Cells, Cultured | 1994 |
Protein kinase C-dependent and -independent pathways mediate epidermal growth factor (EGF) effects in human endometrial adenocarcinoma cell line KLE.
The role of EGF in the proliferation of the poorly differentiated endometrial adenocarcinoma cell line (KLE) was examined. EGF (10 ng/ml) and the tumor promoter, protein kinase C agonist, phorbol 12-myristate 13-acetate (PMA) were potent stimulators (P < 0.01) of DNA synthesis in this cell line as determined by [3H]thymidine incorporation into DNA. Staurosporine, a protein kinase C inhibitor, partially blocked the EGF effects on [3H]thymidine incorporation. Similarly, downregulation of protein kinase C also failed to completely abolish EGF effect on DNA synthesis and cell division. These results suggest that in the KLE cell line EGF stimulation of cell growth is exerted through both protein kinase C-dependent and -independent pathways. Topics: Adenocarcinoma; Alkaloids; Animals; Cell Division; Dose-Response Relationship, Drug; Endometrial Neoplasms; Epidermal Growth Factor; Female; Humans; Mice; Protein Kinase C; Staurosporine; Tetradecanoylphorbol Acetate; Thymidine; Tumor Cells, Cultured | 1994 |
Mitogenic activity of growth factors in the human endometrial adenocarcinoma cell lines HEC-1-A and KLE.
Endometrial adenocarcinoma is the most common gynecologic malignancy occurring in the United States. Evidence is accumulating that links peptide growth factors with malignant proliferation. Epidermal growth factor (EGF) and insulin-like growth factor I (IGF-I) are known mitogens for endometrial adenocarcinoma in vitro. However, the biological activity of other growth factors in this malignancy is unclear. This study was undertaken to determine the influence of growth factors on the mitogenic activity of the human endometrial adenocarcinoma cell lines HEC-1-A and KLE. Incubation with EGF, IGF-I, insulin-like growth factor II (IGF-II), or insulin stimulated a time-dependent mitogenic response in both cell lines, with the peak response occurring at 24 hr for HEC-1-A and 48 hr for KLE. After two doubling intervals, the number of HEC-1-A cells was increased 3.5-fold by EGF (100 ng/ml), 2.7-fold by IGF-I (100 ng/ml), 2.3-fold by IGF-II (100 ng/ml), and 2.2-fold by insulin (1000 ng/ml) when compared to untreated controls (P < 0.05). The number of KLE cells was increased 2.6-fold by EGF (100 ng/ml), 2.3-fold by IGF-I (100 ng/ml), 2.1-fold by IGF-II (100 ng/ml), and 2.0-fold by insulin (1000 ng/ml) when compared to untreated controls (P < 0.05). Similar results were obtained when DNA content was measured. PDGF failed to stimulate any mitogenic response in either cell line at all concentrations tested (0.1-100 ng/ml). These findings suggest that EGF, IGF-I, IGF-II, and insulin may play a regulatory role in the proliferation of endometrial adenocarcinoma. Topics: Adenocarcinoma; Analysis of Variance; DNA, Neoplasm; Endometrial Neoplasms; Epidermal Growth Factor; Female; Growth Substances; Humans; Mitosis; Platelet-Derived Growth Factor; Somatomedins; Thymidine; Time Factors; Tumor Cells, Cultured | 1993 |
Autocrine growth mechanism by transforming growth factor (TGF)-beta 1 and TGF-beta 1-receptor regulation by epidermal growth factor in a human endometrial cancer cell line IK-90.
Transforming growth factor-beta 1 (TGF-beta 1) enhanced cell proliferation in a concentration-dependent manner in a human endometrial cancer cell line, IK-90. Scatchard analysis of TGF-beta 1 receptor in IK-90 cells, using 125I-TGF-beta 1 as a ligand, revealed the presence of a class of high-affinity TGF-beta 1 receptors (2,000 sites per cell, KD = 74pM). Moreover, IK-90 cells produced and secreted TGF-beta 1: TGF-beta 1 messenger RNA was detected at 2.5 and 4.0 kb by Northern-blot analysis using 32P-labeled TGF-beta 1 cDNA as a probe, and TGF-beta 1 activity in conditioned medium by the inhibition of 3H-thymidine uptake into CCl 64 mink lung epithelial cells. We investigated the regulation of TGF-beta 1 receptor by 4 kinds of growth factor: epidermal growth factor (EGF) but not TGF-beta 1, insulin or insulin-like growth factor-1 increased the level of TGF-beta 1 binding sites in a concentration- and time-dependent manner. These findings suggest that TGF-beta 1 may be a potential autocrine growth factor in a human endometrial cancer cell line IK-90 and that this autocrine mechanism may be affected by EGF. Topics: Adult; Cell Division; Culture Media, Serum-Free; DNA, Neoplasm; Endometrial Neoplasms; Epidermal Growth Factor; Female; Humans; Receptors, Cell Surface; Receptors, Transforming Growth Factor beta; Transforming Growth Factor beta; Tumor Cells, Cultured | 1993 |
Regulation of phospholipid turnover by steroid hormones in endometrial carcinoma and breast cancer cells.
To study the early effects of steroid hormones on cells we investigated the influence of the sex steroids and tamoxifen on phospholipid turnover in endometrial carcinoma and breast cancer cells. Studies were performed on 19 human uterine adenocarcinomas and 29 breast cancer tumors. Progesterone in a final concentration of 10(-7) mol/l caused a twofold decrease of 32P incorporation into phospholipids (phosphatidylcholine and phosphoinositides) in 85% of the uterine adenocarcinomas where the progesterone receptor (PR) content was more than 100 nmol/kg and only in 30% of the tumors where the PR content was less than 100 nmol/kg. Treatment of the cells with 10(-8) mol/l 17 beta-estradiol or 10(-8) mol/l epidermal growth factor led to an increase in 32P incorporation into phospholipids. Analysis of the hormonal responsiveness of 29 human breast cancers showed that 17 beta-estradiol increased 32P incorporation into phospholipids in 47% of the tumors where the estradiol receptor (ER) content was more than 10 nmol/kg and in 21% of the receptor-negative tumors (ER < 10 nmol/kg) The results show that phospholipid turnover in uterine and breast cells can be regulated by sex steroids. Treatment of the breast cancer cells with the antiestrogen tamoxifen (10(-6) mol/l) led to an increase of 32P incorporation into phosphoinositides and a decrease of 32P incorporation into phosphatidylcholine. Addition of an activator of protein kinase C, i.e. 2 x 10(-7) mol/l 12-0-tetradecanoylphorbol-13-acetate, weakened the inhibitory effect of tamoxifen on phosphatidylcholine turnover. These findings suggest that tamoxifen action can be mediated via an alteration of the growth signal transducing system. Topics: Adenocarcinoma; Breast Neoplasms; Endometrial Neoplasms; Epidermal Growth Factor; Estradiol; Female; Gonadal Steroid Hormones; Humans; Neoplasms, Hormone-Dependent; Phosphatidylcholines; Phosphatidylinositols; Phospholipids; Progesterone; Tamoxifen; Tetradecanoylphorbol Acetate; Tumor Cells, Cultured | 1993 |
Epidermal growth factor in human breast cancer, endometrial carcinoma and lung cancer. Its relationship to epidermal growth factor receptor, estradiol receptor and tumor TNM.
Epidermal growth factor (EGF) and its receptor (EGFR) were measured in 60 breast cancers (BC), 6 benign mammary tumors (BM), 8 samples of normal breast (NB), 6 endometrial carcinomas (EC) and 30 lung cancers (LC). EGF was measured in plasma, saliva and urine from 20 patients with BC, before and after tumor excision, and in 8 patients with metastatic disease. The median EGF in BM and BC was significantly higher (P < 0.05) than in NB. No significant correlation between EGF and EGFR was found in BC. Neither tumor excision nor the spreading of the disease significantly modified the EGF concentrations in biological fluids. In LC there was an inverse relationship between EGF and EGFR (rs = -0.36; P = 0.09), which disappeared in normal lung. It is concluded that EGF may play a role in malignant transformation; however, the weak correlation between EGF and EGFR lessens the importance of EGF in either autocrine or paracrine stimulation of tumor growth. Topics: Adult; Body Fluids; Breast Neoplasms; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Humans; Lung Neoplasms; Receptors, Estradiol; Saliva | 1993 |
[Cell cycle analysis of endometrial cancer cells in vitro treated with growth factor and steroid hormone].
The aim of this study was to overtake the mechanism of the control system in endometrial cancer cell line in vitro. Ishikawa cell (IK cell) and HEC-1 cell (HEC cell) derived from endometrial cancers were cultured with serum free medium (SFM-101). IK cell possessed Estrogen receptor (ER), Progesterone receptor (PR), Epidermal growth factor (EGF) and its receptor (EGFR). HEC cell had PR, EGF, and EGFR, however HEC cell did not keep ER. EGF stimulated the growth of IK cell, but the growth of HEC cell was not stimulated by EGF. S phase cells were increased by EGF in IK cell, but were not increased by EGF in HEC cell. The growth of IK cell was stimulated significantly by EGF and Estradiol-17 beta (E2) +EGF than control. However, E2+EGF did not stimulate the growth of IK cell than EGF significantly. Danazol (D) and D+EGF inhibited the growth of IK cell significantly than control. S phase cells were decreased by the treatment of D and D+EGF. From our results, EGF stimulated the growth of ER positive endometrial cancer cell, but EGF did not stimulate ER negative endometrial cancer cell. E2+EGF and EGF stimulated the growth of IK cell as a same. However, D inhibited the growth of IK cell that was stimulated by EGF. Topics: Cell Division; Danazol; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Estradiol; Female; Humans; Progesterone; Receptors, Estrogen; Receptors, Progesterone; Tumor Cells, Cultured | 1992 |
Interaction of heparin-binding EGF-like growth factor (HB-EGF) with the epidermal growth factor receptor: modulation by heparin, heparinase, or synthetic heparin-binding HB-EGF fragments.
The binding of heparin-binding EGF-like growth factor (HB-EGF) to the epidermal growth factor (EGF) receptor of human endometrial carcinoma cells was compared to that of EGF using an 125I-EGF radioreceptor assay. The inhibitory effect of HB-EGF on 125I-EGF binding was reversed either in the presence of heparin (but not by chondroitin sulfate) or by pre-treating the cells with heparinase. These treatments did not affect the binding of EGF to its receptor. To map potential regions in the HB-EGF molecule that mediate its heparin-dependent interaction with the EGF receptor, HB-EGF peptides were synthesized that were non-homologous to EGF. Accordingly residues 20-25 and 36-41, but not residues 8-19, of HB-EGF were found to be (i) heparin-binding and (ii) modulators of HB-EGF (but not of EGF) binding to the EGF receptor. Topics: Amino Acid Sequence; Binding, Competitive; Chromatography, High Pressure Liquid; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Female; Heparin; Heparin Lyase; Heparin-binding EGF-like Growth Factor; Humans; Intercellular Signaling Peptides and Proteins; Molecular Sequence Data; Peptide Fragments; Polysaccharide-Lyases; Radioligand Assay; Tumor Cells, Cultured | 1992 |
[In vitro study on the effect of sex steroid and growth factor on growth and laminin, collagen IV, and tissue plasminogen activator production of normal endometrial cells and endometrial cancer cells in culture].
The mechanism of the invasion and proliferation of endometrial cancer is closely related to interactions between the endometrial glands and stroma. In this study, we examined the biological role of sex steroids (estradiol; E2, progesterone; P) and growth factors (epidermal growth factor; EGF, transforming growth factor-beta; TGF-beta) on cell growth and laminin, collagen IV and tissue plasminogen activator (t-PA) production of normal endometrial cells and endometrial cancer cells in culture. Normal endometrial gland cells and stromal cells, and endometrial cancer cell lines (Ishikawa, OMC-2) were used. E2, P, EGF and TGF-beta were added to the culture in physiological concentrations. The growth of normal endometrial gland cells was promoted by E2 and EGF, whereas that of Ishikawa cells and OMC-2 cells was promoted by EGF. E2 enhanced the effects of EGF in normal endometrial gland cells. The growth of normal endometrial stromal cells was not affected by them. OMC-2 was inhibited by anti-EGF receptor antibody. On the other hand, the production of laminin and collagen IV of these cultured cells was inhibited by EGF and promoted by TGF-beta, whereas that of t-PA was promoted by EGF and inhibited by TGF-beta. These results suggest that the growth of normal endometrial gland cells with estrogen receptor (ER) is controlled by both E2 and EGF, whereas that of endometrial cancer cells is affected only by EGF, and those cells without ER depend particularly on the autocrine growth mechanism of EGF.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Cell Division; Cells, Cultured; Collagen; Endometrial Neoplasms; Endometrium; Epidermal Growth Factor; Estradiol; Female; Humans; Laminin; Progesterone; Tissue Plasminogen Activator; Transforming Growth Factor beta; Tumor Cells, Cultured | 1992 |
Effects of transforming growth factors and regulation of their mRNA levels in two human endometrial adenocarcinoma cell lines.
The effects of the transforming growth factor-beta 1 (TGF-beta 1) and epidermal growth factor (EGF) on the growth of cells from 2 endometrial cancer lines, Ishikawa and HEC-50 were evaluated by measuring rates of DNA synthesis and changes in cell numbers during culture. EGF at 17 and 1.7 nM concentrations consistently enhanced HEC-50 cell proliferation. TGF-beta 1 inhibited Ishikawa cell proliferation but, unexpectedly for epithelium-derived cells, stimulated HEC-50 cell growth. This effect is of interest as it indicates that endometrial cells can acquire an altered responsiveness to a growth inhibitor during the process of malignant transformation. Northern blot analyses showed expression of TGF-alpha, TGF-beta 1 and EGF receptors mRNA in both cell lines. Neither estradiol (E2) nor 4-hydroxytamoxifen (OHTam) affected mRNA levels for either TGF-alpha or TGF-beta in HEC-50 cells, a line unresponsive to E2 for proliferation. In Ishikawa cells, previously shown to respond to both E2 and OHTam by increasing proliferation rates, E2 increased TGF-alpha mRNA and reduced TGF-beta mRNA levels. OHTam lowered the levels of both mRNA species, although the effect was greater on TGF-beta than TGF-alpha mRNA. These data are consistent with, but do not prove, the existence of a possible autocrine regulation by TGF-alpha and TGF-beta of human cancer cell proliferation, which might be under E2 influence in Ishikawa cells. Topics: Adenocarcinoma; Blotting, Northern; Cell Division; DNA; Endometrial Neoplasms; Epidermal Growth Factor; Estrogen Antagonists; Female; Humans; RNA, Messenger; Tamoxifen; Transforming Growth Factor alpha; Transforming Growth Factor beta; Tumor Cells, Cultured | 1992 |
Endothelin-1 gene expression and biosynthesis in human endometrial HEC-1A cancer cells.
In this study, evidence was obtained that endothelin-1 (ET-1) is produced by an established endometrial cancer (HEC-1A) cell line. PreproET-1 mRNA is present in HEC-1A cells, and immunoreactive endothelin is secreted into the medium of these cells maintained in culture. Cycloheximide treatment of these cells caused superinduction of preproET-1 mRNA. Transforming growth factor-beta acts in these cells to increase the levels of preproET-1 mRNA. This effect of transforming growth factor-beta on preproET-1 mRNA accumulation was accompanied by an increase in the amount of immunoreactive endothelin secreted into the culture medium. ET-1, added to the culture medium, did not act as a mitogen in HEC-1A cells. We speculate that ET-1 (which is known to stimulate fibroblast proliferation) produced by endometrial adenocarcinoma cells may participate in the angiogenic process that occurs during the establishment of this carcinoma in vivo. Topics: Adenocarcinoma; DNA Replication; DNA, Neoplasm; Endometrial Neoplasms; Endothelin-1; Endothelins; Epidermal Growth Factor; Female; Fibroblast Growth Factors; Gene Expression; Humans; Insulin; Interleukin-1; Kinetics; Platelet-Derived Growth Factor; Protein Precursors; RNA, Messenger; Transforming Growth Factor beta | 1992 |
[Responsiveness and mechanisms of action of steroid hormones in human endometrial adenocarcinoma cells].
The responsiveness and action mechanisms of steroid hormones and epidermal growth factor on human endometrial carcinoma cells are analyzed by using in vitro culture system. 1) The Ishikawa cells, derived from a well differentiated endometrial adenocarcinoma and possess ER and PR, are shown to respond to estrogens by increasing a variety of parameters, viz cell proliferation, PR levels, ALP and DNA polymerase activities. 2) ER and PR of those cells are localized in the nuclei by immunocytochemical staining using the monoclonal antibodies against to ER and PR, confirming the correctness of Gorski and Greene's one step theory involving the action mechanisms of steroid hormones. 3) Progestins reduced the ER level and stimulate E2DH activities and glycogen content, which are completely abolished by anti-progestin (RU486), suggesting that PR of those cells should be functional. 4) These responses to steroid hormones of Ishikawa cells are synergistically enhanced or appeared earlier by addition of EGF. 5) The main metabolite of E2 incubated with Ishikawa cells is E2-3-sulfate instead of E1, indicate that the higher estrogenic status may be persisted in endometrial cancer tissues. Topics: Adenocarcinoma; Alkaline Phosphatase; Cell Division; DNA Polymerase II; Endometrial Neoplasms; Epidermal Growth Factor; ErbB Receptors; Estradiol Dehydrogenases; Estrogens; Female; Glycogen; Humans; Progesterone; Receptors, Estrogen; Receptors, Progesterone; Tumor Cells, Cultured | 1989 |