Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Cancer ; 123(3): 616-22, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18498128

ABSTRACT

Usual type VIN is a premalignant disorder caused by persistent HPV infection. High prevalence of VIN in immuno-suppressed women suggests that a good innate and adaptive immune response is important for defense against HPV. Here, we explored expression levels of chemokines and related these to the presence or absence of immuno-competent cells (dendritic and T-cells) in affected (HPV-positive VIN) and non-affected (HPV-negative) vulvar tissues from the same patients. Combining microarray data with quantitative real-time RT-PCR, it was observed that several important chemokines were differentially expressed between VIN and control samples (up-regulation of IL8, CXCL10, CCL20 and CCL22 and down-regulation of CXCL12, CCL21 and CCL14). Furthermore, an increased number of mature dendritic cells (CD208+) seemed to be bottled up in the dermis, and although a T-cell response (increased CD4+ and CD8+ cells) was observed in VIN, a much larger response is required to clear the infection. In summary, it seems that most mature dendritic cells do not receive the proper chemokine signal for migration and will stay in the dermis, not able to present viral antigen to naive T-cells in the lymph node. Consequently the adaptive immune response diminishes, resulting in a persistent HPV infection with increased risk for neoplasia.


Subject(s)
Alphapapillomavirus , Chemokines/metabolism , Dendritic Cells/immunology , Immunocompromised Host , Papillomavirus Infections/complications , T-Lymphocytes/immunology , Tumor Virus Infections/complications , Uterine Cervical Dysplasia/immunology , Uterine Cervical Neoplasms/immunology , Adult , Chemokines/genetics , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Microarray Analysis , Middle Aged , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology , Up-Regulation , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/virology
2.
Reprod Sci ; 14(7): 646-54, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18000226

ABSTRACT

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


Subject(s)
Endometrium/metabolism , Estradiol/pharmacology , Estrogens/physiology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Growth Factor/metabolism , Tamoxifen/pharmacology , Cell Line, Tumor , Endometrium/drug effects , ErbB Receptors/metabolism , Estrogens/genetics , Estrogens/pharmacology , Female , Gene Expression Profiling , Humans , Insulin-Like Growth Factor I/metabolism , Multigene Family , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-akt/genetics , Receptors, Growth Factor/genetics , Signal Transduction
3.
J Soc Gynecol Investig ; 12(7): e55-67, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16202921

ABSTRACT

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


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Endometrial Neoplasms/pathology , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Glycoproteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Amphiregulin , EGF Family of Proteins , Estradiol/pharmacology , Estradiol/physiology , Female , Fulvestrant , Gene Expression Profiling , Humans , Transcription, Genetic , Tumor Cells, Cultured
4.
J Soc Gynecol Investig ; 12(4): 285-92, 2005 May.
Article in English | MEDLINE | ID: mdl-15866122

ABSTRACT

OBJECTIVE: Progesterone plays an important role in controlling proliferation and differentiation of the human endometrium. Because there are two progesterone receptor isoforms (PRA and PRB), it was important to generate tools to be able to study the role of these two progesterone receptors separately. METHODS: Using stable transfection techniques, both human progesterone receptor isoforms (hPRA and hPRB) were reintroduced into a hPR-negative subclone of the well-differentiated endometrial cancer cell line Ishikawa. Several Ishikawa subcell lines were constructed, each expressing different levels of hPRA, hPRB, or hPRA and hPRB, respectively. RESULTS: These Ishikawa subcell lines showed a marked progesterone-induced growth inhibition with induction of apoptosis after long-term culture in the presence of hormone. Upon measuring gene regulation, a clear difference in regulation of expression of the selected genes by progesterone treatment was observed between the PRA-, PRB-, or PRA/B-expressing cell lines. Integrin beta4 (ITGB4) was only regulated in PRA-expressing cells; amphiregulin was highly regulated in PRB-expressing cells; insulin-like growth factor binding protein 3 (IGFBP3) was only regulated in PRB- and PRA/B-expressing cells; and metallothionein 1L (MT1L) was highly regulated in PRA/B-expressing cells. Interestingly, based on literature data, these genes can be implicated in induction of apoptosis, but are modulated here in such a way that suggests induction of resistance against apoptosis. CONCLUSION: Reintroduction of PRs into Ishikawa cells rescued progesterone responsiveness in these cells. Furthermore, using these human endometrial cancer subcell lines, clear and distinct functional differences between the PR isoforms were observed.


Subject(s)
Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Progesterone/physiology , Receptors, Progesterone/biosynthesis , Apoptosis , Cell Differentiation , Cell Proliferation , Female , Gene Expression Profiling , Humans , Protein Isoforms , Transfection , Tumor Cells, Cultured
5.
Clin Cancer Res ; 9(11): 4190-9, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14519645

ABSTRACT

PURPOSE: In endometrial cancer, loss of progesterone receptors (PR) is associated with more advanced disease. This study aimed to investigate the mechanism of action of progesterone and the loss of its receptors (PRA and PRB) in development of endometrial cancer. EXPERIMENTAL DESIGN: A 9600-cDNA microarray analysis was performed to study regulation of gene expression in the human endometrial cancer subcell line Ishikawa PRAB-36 by the progestagen medroxy progesterone acetate (MPA). Five MPA-regulated genes were selected for additional investigation. Expression of these genes was studied by Northern blot and by immunohistochemistry in Ishikawa subcell lines expressing different PR isoforms. Additionally, endometrial cancer tissue samples were immunohistochemically stained to study the in vivo protein expression of the selected genes. RESULTS: In the PRAB-36 cell line, MPA was found to regulate the expression of a number of invasion- and metastasis-related genes. On additional investigation of five of these genes (CD44, CSPG/Versican, Tenascin-C, Fibronectin-1, and Integrin-beta 1), it was observed that expression and progesterone regulation of expression of these genes varied in subcell lines expressing different PR isoforms. Furthermore, in advanced endometrial cancer, it was shown that loss of expression of both PR and E-cadherin was associated with increased expression CD44 and CSPG/Versican. CONCLUSION: The present study shows that progestagens exert a modulatory effect on the expression of genes involved in tumor cell invasion. As a consequence, loss of PR expression in human endometrial cancer may lead to development of a more invasive phenotype of the respective tumor.


Subject(s)
Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Receptors, Progesterone/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Line, Tumor , Chondroitin Sulfate Proteoglycans/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Receptors/genetics , Lectins, C-Type , Medroxyprogesterone Acetate/pharmacology , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Tenascin/genetics , Versicans
6.
J Soc Gynecol Investig ; 10(1): 49-57, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12517594

ABSTRACT

To study the functional differences between the two progesterone receptor isoforms (hPRA and hPRB) in human endometrial cancer, two new endometrial carcinoma cell lines were created-one expressing hPRA and one expressing hPRB.A well-differentiated, hPR-negative Ishikawa cell line was stably transfected with either hPRA or hPRB cDNA. Transfected cells were selected, and two cell lines expressing approximately equal amounts of receptor were isolated-one expressing hPRA (PRA-14) and one expressing hPRB (PRB-59). Cell growth experiments revealed a growth-inhibitory response to progestins (MPA and R5020) in the PRB-59 cells but not in the PRA-14 cells. Differences in expression of genes targeted by the two isoforms were studied using a cDNA expression array technique. A different set of genes appeared to be progesterone regulated in the PRA-14 cells than in the PRB-59 cells. None of the genes were regulated by both hPRA and hPRB. Insulin-like growth factor binding protein 3 expression was studied in more detail as an example of a gene regulated in PRB-59 cells but not in PRA-14 cells. We established a new model to study functional differences between the two hPR isoforms in human endometrial carcinoma cells. This model revealed distinctive differences in target gene regulation between the two hPR isoforms. Moreover, antiproliferative actions of progesterone on human endometrial cancer cells could be observed only in the PRB-expressing cell line.


Subject(s)
Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Receptors, Progesterone/physiology , Cell Division/drug effects , Female , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medroxyprogesterone Acetate/pharmacology , Progesterone/pharmacology , Progesterone Congeners/pharmacology , Promegestone/pharmacology , Receptors, Progesterone/genetics , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...